<<

Facilitating End-to-End Development of Individualized Therapeutics Public Workshop U.S. Food and Drug Administration Center for Biologics Evaluation and Research (CBER)

FDA White Oak Campus 10903 New Hampshire Ave. Bldg. 31 Conference Center Great Room Silver Spring, MD 20993

March 3, 2020

This transcript appears as received from the commercial transcribing service after inclusion of minor corrections to typographical and factual errors recommended by the DFO

2

ATTENDEES

Albert B. Seymour, PhD Homology Medicines, Inc.

Alison Bateman-House, PhD, MPH, MA NYU Grossman School of Medicine

Donald B. Kohn, MD UCLA

Guangping Gao, PhD University of Massachusetts

Jason J. Gill, PhD Texas A&M University

Jill A. Wood Phoenix Nest

J. Keith Joung, MD, PhD Massachusetts General Hospital

Malachi Griffith, PhD Washington University School of Medicine

Philip John (PJ) Brooks, PhD National Institutes of Health

Robert T. (Chip) Schooley, MD University of California San Diego Center for Biologics Evaluation and Research Gopa Raychaudhuri, PhD (CBER) Center for Biologics Evaluation and Research Peter Marks, MD, PhD (CBER) Center for Biologics Evaluation and Research Zenobia Taraporewala, PhD (CBER) Center for Biologics Evaluation and Research Roger Plaut, PhD (CBER) Center for Biologics Evaluation and Research Anita Richardson, MAS (CBER) Center for Biologics Evaluation and Research Sandhya Sanduja, PhD (CBER) Center for Biologics Evaluation and Research Zuben Sauna, PhD (CBER) Center for Biologics Evaluation and Research Rebecca Reindel, MD (CBER) Center for Biologics Evaluation and Research Larissa Lapteva, MD (CBER) Center for Biologics Evaluation and Research Zhenzhen Xu, PhD (CBER)

www.transcriptionetc.com

3

Center for Biologics Evaluation and Research Celia Witten, PhD, MD (CBER) Center for Biologics Evaluation and Research Capt. Julie Vaillancourt (CBER) Stephen Aldrich MyCancerDB

Joe Campbell NIAID

Matt Kelly Sarepta Therapeutics

Sharon Hesterlee Muscular Dystrophy Association Advanced Regenerative Manufacturing Richard McFarland Institute Karen Walker Genentech

Jessica Adomako Genentech

Marilyn Howard University of Pennsylvania Center for Biologics Evaluation and Research Carolyn Wilson, PhD (CBER) Lea Witkowsky Innovative Genomics Institute

Aron Stein Sangamo Therapeutics

Lorraine McLellan Audience Questioner

Lynne McGrath Audience Questioner

Neil Thakur ALS Association

Rich Horgan Cure Rare Disease

Anita Nosratieh FasterCures

Lauren Black Charles River Laboratories

www.transcriptionetc.com

4

TABLE OF CONTENTS

INTRODUCTION: DR. GOPA RAYCHAUDHURI ...... 6 SPEAKER: DR. PETER MARKS ...... 11 SESSION 1: MANUFACTURING ...... 22 SESSION 1 MODERATOR INTRODUCTION: DR. ZENOBIA TARAPOREWALA ...... 22 CHALLENGES AND OPPORTUNITIES IN DEVELOPMENT AND MANUFACTURING OF INDIVIDUALIZED THERAPEUTICS WITH AAV VECTOR-BASED THERAPIES - DR. GUANGPING GAO ...... 30 DEVELOPMENT OF PHAGE THERAPY: PERSONALIZED MEDICINE AND INDIVIDUALIZED THERAPEUTICS - DR. JASON J. GILL ...... 51 PANEL SESSION WITH Q&A ...... 74 [BREAK] ...... 96 SESSION 2: TOOLS FOR SAFETY TESTING AND DEVELOPMENT ...... 96 SESSION 2 MODERATOR INTRODUCTION: DR. SANDHYA SANDUJA (CBER) ...... 96 PRECLINICAL APPROACHES/CHALLENGES IN DEVELOPMENT OF INDIVIDUALIZED THERAPEUTICS - DR. ALBERT B. SEYMOUR ...... 104 BIOINFORMATICS TOOLS FOR DEVELOPMENT, ANALYSIS & PRECLININCAL TESTING OF INDIVIDUALIZED THERAPEUTICS - DR. MALACHI GRIFFITH ...... 128 DEFINING OFF-TARGET EFFECTS OF GENE EDITING TECHNOLOGIES - DR. J. KEITH JOUNG ...... 150 PANEL DISCUSSION WITH Q&A ...... 174 SESSION 3: CLINICAL ...... 200 SESSION 3 MODERATOR INTRODUCTION: DR. REBECCA REINDEL ...... 201 OPPORTUNITIES AND CHALLENGES IN THE CLINICAL DEVELOPMENT OF BACTERIOPHAGE THERAPEUTICS - DR. ROBERT T. SCHOOLEY...... 209 CHALLENGES TO DEVELOPING INDIVIDUALIZED STEM CELL GENE THERAPIES - DR. DONALD B. KOHN ...... 230 PANEL SESSION WITH Q&A ...... 253 SESSION 4: PRODUCTS TO PATIENTS ...... 276 SESSION 4 MODERATOR INTRODUCTION: DR. CELIA WITTEN ...... 276 THE TRIALS AND TRIBULATIONS OF DRIVING A TREATMENT FOR AN UBER-RARE DISEASE TO THE CLINIC AND BEYOND-A PARENT’S PERSPECTIVE - MS. JILL A. WOOD ...... 286 ETHICAL ISSUES IN PRODUCT DEVELOPMENT AND SUSTAINABILITY FOR INDIVIDUALIZED THERAPIES-DR. ALISON BATEMAN-HOUSE ...... 308

www.transcriptionetc.com

5

BEYOND ‘ONE DISEASE AT A TIME:’ ACCELERATING CLINICAL TRIALS OF GENETIC THERAPIES BY GROUPING RARE DISEASE PATIENTS ACCORDING TO UNDERLYING DISEASE MECHANISM-DR. PHILIP J. BROOKS ...... 330 PANEL DISCUSSION WITH Q&A ...... 349 WRAP UP AND CLOSING REMARKS: DR. PETER MARKS ...... 378

www.transcriptionetc.com

6

1 INTRODUCTION: DR. GOPA RAYCHAUDHURI

2

3 DR. RAYCHAUDHURI: Good morning, everyone, and

4 welcome to the CBER workshop on facilitation and

5 development of individualized therapeutics. My name is

6 Gopa Raychaudhuri. I'm a senior scientist in the

7 Office of the Director at CBER, and I coordinate and

8 oversee CBER's individualized therapeutics program.

9 It is my distinct pleasure to welcome all the

10 stakeholders who are here today, including patients,

11 family members, patient advocacy organizations,

12 healthcare professionals, and individuals from

13 nonprofit organizations, academia, industry, and

14 government. In addition, I would like to thank

15 everyone who's participating in today's proceedings via

16 webcast. We appreciate you taking the time to join and

17 contribute online.

18 Before we begin today's proceedings, I have a

19 few general announcements. First, please silence your

20 cell phones and any mobile devices. If you haven't

21 done so already, we ask that all attendees sign in at

www.transcriptionetc.com

7

1 the registration tables just outside this meeting room.

2 If you would like to preorder lunch, you can

3 do so at the food kiosk that's just outside the

4 conference room. Lunch must be preordered by 9:30. If

5 you decide not to preorder, you may purchase snacks,

6 sandwiches, and other food items a la carte at the

7 kiosk. And that's open until 5:00 today.

8 This meeting is being transcribed, and a live

9 webcast is being recorded. There also is an official

10 photographer, as you can see, who will be taking photos

11 during the course of the workshop. For any urgent

12 issues, please speak to the registration staff that are

13 just outside or any FDA staff member with a tag, and we

14 will be able to assist.

15 I'd like to open today's workshop by defining

16 what we mean by individualized therapeutics and why we

17 need to think differently about development, licensure,

18 and access to these products. Out of thousands of

19 rare, hereditary, and acquired diseases, there are

20 hundreds of disorders affecting one or a small group of

21 people each year that could be addressed with

www.transcriptionetc.com

8

1 individualized or bespoke therapies. These therapies

2 are based on engineering a product aimed at the

3 specific mechanism underlying a patent's, or a small

4 group of patients', illness. That is the therapeutic

5 product is engineered specifically for a given patient

6 or small group of patients.

7 Examples include gene therapy or gene editing

8 for monogenic diseases, antisense oligonucleotides, or

9 genetically-engineered phages for multidrug-resistant

10 infection. Today's workshop will focus on

11 individualized therapeutic products regulated by CBER,

12 specifically, gene therapies and phage therapies. But

13 we work in close collaboration with our colleagues in

14 CDER, who are very active in this space to facilitate

15 development of antisense oligonucleotide products for

16 patients with rare diseases.

17 This slide shows the traditional regulatory

18 pathway, from discovery to licensure and post-licensure

19 monitoring, for biologics and other medical products

20 regulated by FDA. Individualized therapeutic products

21 present unique challenges because they do not fit the

www.transcriptionetc.com

9

1 traditional paradigm for manufacturing and clinical

2 development. This requires that we think outside the

3 box, to establish an evidence-based, clear, and

4 practical pathway for development, regulation, and

5 access for patients to these products, while assuring

6 that the standards for quality, safety, and efficacy

7 are maintained.

8 During the course of this workshop, you will

9 hear about existing challenges and potential solutions

10 to adapt the current process to meet the need and

11 opportunities for stakeholder collaboration to move

12 this field of work forward. This slide shows the large

13 number and wide range of stakeholders that play a

14 critical role in this field of work. As a community

15 working towards a common goal, each stakeholder brings

16 valuable perspective, knowledge, skills, and resources

17 to the effort. We are pleased that so many stakeholder

18 groups are participating in this workshop. And we look

19 forward to your contribution during today's

20 discussions.

21 The workshop is divided into four sessions.

www.transcriptionetc.com

10

1 The first is on manufacturing. The second session

2 focuses on nonclinical development and tools for safety

3 testing. The third is on clinical development. And I

4 should note that there is one change in the program in

5 the clinical section. Unfortunately, Dr. Kohn is not

6 able to be here in person because of flight delays

7 yesterday. But he has kindly agreed to join us

8 remotely and will be giving his presentation remotely.

9 And the fourth session will focus on what is the

10 ultimate objective, which is getting products to

11 patients in an efficient and sustainable way and the

12 critical role of partnerships and collaborations to

13 make this a reality.

14 Each session will start with a short

15 introduction by one of my FDA colleagues, who will

16 serve as the moderator for the session. That will be

17 followed by two or three 20-minute presentations by

18 external experts. And we are very happy to have each

19 of them here today.

20 Following the presentations, there will be a

21 panel discussion, where we will open the floor for

www.transcriptionetc.com

11

1 comments and questions for the speakers and panelists.

2 Online participants will have an opportunity to write

3 in their questions. This is an important part of

4 today's workshop because one of our main objectives is

5 to hear from you, the stakeholders, on what we can do

6 as individual organizations and collectively as a

7 community to facilitate end to end development and

8 timely and sustainable access to these important

9 products for the patients who need them.

10 And with that, I would like -- it's my

11 pleasure to introduce Dr. Peter Marks. Dr. Marks is

12 the Director for the Center for Biologics Evaluation

13 and Research at FDA. And he will give a few

14 introductory remarks on the current state, and he will

15 set the stage for today's discussion. Dr. Marks.

16 SPEAKER: DR. PETER MARKS

17

18 DR. MARKS: Thanks very much. Good morning.

19 And thanks everyone in the room for coming today. And

20 thank you all online for listening in.

21 I just want to spend a couple minutes setting

www.transcriptionetc.com

12

1 the stage for what leads us to be here today. And one

2 of those things is just to back up and say where we've

3 come over the past few decades. At the end of the last

4 millennium, we were in this era of personalized

5 medicine, where, increasingly, we were beginning to

6 understand characteristics of people's disease that led

7 us to choose medicines off the shelf that uniquely

8 address their diseases, whether that be for cancers,

9 infectious diseases, rheumatologic diseases.

10 And so this concept of personalized medicines,

11 where you find the right drug on the shelf to treat the

12 patient, became prevalent. But we're now moving into a

13 different era. And that's an era where we understand

14 the molecular defects at the level of the genome, and

15 we actually are needing to create the right drug to

16 treat the patient. We don't have things right off the

17 shelf. And so what we're really moving into is an era

18 of individualized medicine.

19 And yes, one could say that this is a semantic

20 difference, but there is a difference here in how we as

21 regulators, and I think how we as a field in general,

www.transcriptionetc.com

13

1 deal with this. And I just want to say that, as you

2 start to cogitate over this some, you realize that

3 individualized medicine actually breaks down further

4 into products that are essentially customized products

5 and products that are created. The customized products

6 are one where you could imagine they have the same

7 indication and the same mode of action, but there's

8 something different about each one that's unique for an

9 individual.

10 One can imagine that if one is making

11 dendritic cells against some cancer that a patient has,

12 some particular cancer -- it could be a lung cancer or

13 some other cancer. But if you have different product,

14 because it's got different peptides on it, you could

15 have a lot of different ones with some unique attribute

16 on some basic product. And those are customized.

17 On the other hand, there are products which

18 are going to be for different indications, different

19 lysosomal storage disorders, which may be very similar

20 in that they have the same backbone vector, if they're

21 a gene therapy, but they'll have different inserts.

www.transcriptionetc.com

14

1 And so I think this concept of customized and created

2 products is something that we'll hear more about today,

3 but it is something that we'll think about going

4 forward. And there probably are some regulatory

5 distinctions between these two classes.

6 Just to make this point a little further,

7 personalized medicine is a little bit like ready-to-

8 wear. You find stuff on the shelf that kind of fits

9 you, and it just is -- it's there. It's like medicines

10 off the shelf that you find that fit.

11 On the other hand, this concept of customized

12 products probably has the analogy of something that's

13 made to measure. Made to measure, for those of you who

14 are not familiar with tailoring -- and I'm not one to

15 talk because I'm not one for sartorial splendor -- made

16 to measure is actually not a custom-made suit, a made

17 to measure suit is one in which the fabric's been all

18 cut. It's just the seams are left open so that they

19 can be stitched closed once they adjust it to your fit.

20 And that's kind of the analogy to customized products.

21 On the other hand, there are these created

www.transcriptionetc.com

15

1 products, which is more like a bespoke suit. A bespoke

2 suit is somebody -- you choose the yards of cloth and

3 someone cuts them and puts them together. And so this

4 is kind of the analogy here I'd like to use. It's not

5 perfect, but it does give us the idea of what we're

6 dealing with.

7 Now, there are some challenges that go with

8 these individualized therapies. And that's what we're

9 here to talk about today. And we have sessions to talk

10 about manufacturing, nonclinical development, clinical

11 development, and product access. And we certainly

12 don't have the answers for these. But I think I'd like

13 to go through and kind of pose some of the questions.

14 Just to show you on the manufacturing side,

15 manufacturing in this area is really a challenge. In

16 the field of gene therapy, we have moved so fast in the

17 science we're now on to second and third generation of

18 genome editing constructs. We've moved on past the

19 initial generation of CRISPR to new novel genome

20 editors. That being said, we're still making vectors

21 much the same way that we made them at the turn of the

www.transcriptionetc.com

16

1 millennium. And we really need to figure out how to

2 move that forward.

3 The reason for that is that, right now, we

4 have one sweet spot, which is the grande-size cup in

5 the middle, where commercial viability relies on

6 products that are going to treat between a hundred and

7 maybe a thousand or a few thousand people. If one

8 wanted to treat very large numbers of patients --

9 that's something for another day's talk and another

10 day's meeting -- the technology is simply not there.

11 One would need essentially to fill Lake Erie as a

12 bioreactor to make enough vector to treat thousands of

13 people. That's just not where we are. We're going to

14 have to find more efficient ways to do that.

15 On the other hand, what we are here to talk

16 about today is how do we deal with making these

17 products for small numbers of individuals. And that's

18 a challenge because the setup costs currently to make a

19 gene therapy for 20 people are very similar to the

20 setup costs to make a gene therapy for 200 people, in

21 terms of a commercial process. So that's something we

www.transcriptionetc.com

17

1 have to really think about because it's limiting the

2 ability to get access to these products.

3 Nonclinical development is also a challenge

4 because animal models may be much less than ideal here.

5 And that's particularly true when we start to think of

6 genome editing because you might want to do toxicity

7 studies of a genome editor. You might want to

8 understand off-target effects on a genome editor.

9 But last I looked, although some people think

10 I look a little like a mouse, my genome does not look

11 exactly like a mouse. And it's a real problem.

12 Despite the fact that there's a fair amount of

13 identity, there's enough difference that you wouldn't

14 want to trust a mouse model for a genome editor with

15 the guide. So we may need to think about new model

16 systems, such as how can human organoid systems,

17 humanized mice, other models, help in this area for

18 safety testing.

19 For clinical development, I think we really

20 are going to have to think very novel-ly about how we

21 deal with very small populations. We're going to have

www.transcriptionetc.com

18

1 to think about how we document disease, the natural

2 history of disease, or collect baseline data so that,

3 when we actually have small numbers of patients

4 treated, we can see that there's some difference from

5 that baseline.

6 And once we start to treat patients, can we

7 find some way of using some type of continuous

8 reassessment model or some other statistical method, a

9 Bayesian or other design, to see if there's some point

10 at which one crosses where one declares victory that

11 you actually have an effective product, rather than

12 developing a sample size and running a traditional

13 clinical trial. And very importantly, we have to deal

14 with product access. How do we deal with the fact

15 that, for some of these product, once you've even

16 gotten them through development, how do you make sure

17 you have continued access to them given the complexity

18 and the cost of providing that access?

19 We already know what can happen. One of the

20 gene therapies that was approved in Europe is now no

21 longer on the market. Now, obviously, it can be made

www.transcriptionetc.com

19

1 available in other ways. But it shows you what can

2 happen here if we don't think about access and we don't

3 think about the costs and the complexity of these

4 products.

5 And so one of the things we've been thinking

6 about here is whether public-private partnerships could

7 at least help access in the United States and perhaps

8 even globally through the streamlined production of

9 products. Because when you think about it, if we're

10 making products for very small numbers of patients with

11 rare diseases, we shouldn't reinvent the wheel

12 globally. There shouldn't be initiative here in the

13 United States, initiative in Europe, initiative in

14 Asia. It probably would be better to have everyone

15 contributing to the effort.

16 So the idea here of a gene therapy public-

17 private partnership would be to try to drive down the

18 time it takes to get these products to patients by

19 using reproduceable processes or leveraging data to

20 make things happen. Because, typically, when

21 investigators, individual investigators develop a gene

www.transcriptionetc.com

20

1 therapy, they go through all the normal stages of drug

2 development, including submitting an IND, doing the

3 necessary tox studies, doing the manufacturing, which

4 oftentimes they have to learn to do from scratch. And

5 that whole process takes four to eight years often on

6 average.

7 One could imagine that if one was able to

8 leverage manufacturing processes, use templated INDs,

9 templated protocols -- obviously, it would have to be

10 customized somewhat -- you could potentially shorten

11 this a fair amount. And those -- that shortening of

12 years means a lot. For patients, when you talk to

13 parents of children with rare diseases, years matter, a

14 few years matter.

15 So our goal of this workshop is really to

16 think about where the opportunities are to adapt

17 processes from beginning to end. You see the

18 traditional process here. No one is suggesting that,

19 for these products, we're going to have traditional

20 phase one, two, three development.

21 The question is, where can we go with these to

www.transcriptionetc.com

21

1 end up at the end of the day with products that are

2 both safe and effective? Because I think, in the

3 process of trying to develop this paradigm, we want to

4 maintain the ability to know that, at the end of the

5 day, what we're providing to patients is something that

6 is similar in nature to the gold standard that we

7 provide patients with now with approved products. And

8 with that, I will turn it back over to Gopa.

9 But before I do, I just want to -- I just

10 nearly forgot one thing. I mean to have a slide here.

11 But I want to just take the moment to thank both Dr.

12 Raychaudhuri, as well as Leslie Haynes, for tremendous

13 work putting together this workshop.

14 There is a -- there are others as well from

15 our office who spent a lot of time taking care of

16 logistics of travel, making sure that things run well.

17 So thank you so much. And with that, I will wish

18 everyone a wonderful day. We look forward to the

19 discussions. Thank you.

20 DR. RAYCHAUDHURI: Thank you, Dr. Marks, for

21 that overview and vision of approaches and

www.transcriptionetc.com

22

1 opportunities that we can think about to advance

2 development and access to individualized therapeutic

3 products.

4 So we will begin the program now with Session

5 1. I would like to introduce Dr. Zenobia Taraporewala.

6 Dr. Taraporewala is a CMC reviewer, and Acting Team

7 Lead for the Gene Therapy Branch in the Division of

8 Cellular and Gene Therapies in the Office of Tissues

9 and Advanced Therapies at CBER. Dr. Taraporewala will

10 be the moderator for Session 1, which is on

11 manufacturing challenges and opportunities for gene

12 therapy and phage therapy products. Dr. Taraporewala?

13 SESSION 1: MANUFACTURING

14 SESSION 1 MODERATOR INTRODUCTION: DR. ZENOBIA

15 TARAPOREWALA

16 DR. TARAPOREWALA: Good morning. All right.

17 So let's get started. As Gopa said, I am Zenobia

18 Taraperwala, and I am from the Office of Tissues and

19 Advanced Therapies, commonly referred to as OTAT in

20 CBER. And I will be the moderator for the session.

21 We have two very distinguished speakers in the

www.transcriptionetc.com

23

1 session, Dr. Guangping Gao from University of

2 Massachusetts Medical School and Dr. Jason Gill from

3 Texas A&M University. Following the two presentations,

4 we will have a panel discussion. And the panel will

5 include speakers from this session and CBER

6 representatives from the Office of OVRR -- from OVRR

7 the Office of Vaccine Research Review, Dr. Roger Plaut,

8 and from the Office of OCBQ, which is Ms. Anita

9 Richardson. And during the panel, we will field

10 questions from the audience and from those attending

11 online.

12 So this is advancing but that is not. Okay.

13 Thank you. All right. So this slide illustrates the

14 manufacturing development, where manufacturing

15 feasibility is assessed, and analytical tests are -- so

16 this basically gives you a manufacturing development

17 paradigm that is currently followed for biologics and

18 drugs in general.

19 It starts with the discovery and the

20 preclinical stage where manufacturing feasibility is

21 assessed and analytical tests that are suitable for

www.transcriptionetc.com

24

1 release of research and developmental lots and IND-

2 enabling lots are developed. That manufacturing

3 experience is then leveraged for the manufacturing of

4 clinical lots under GMP suitable for early phase

5 studies. And with the release of multiple clinical

6 lots, there is greater understanding of the variables

7 in the starting and the raw materials and for the

8 process on analytical assays used in release testing,

9 all that are critical to ensure lot-to-lot consistency.

10 In late phase studies, the analyticals needed

11 for thorough product characterization and in-process

12 testing to meet the future demands of commercial

13 process are put in place. Finally, for licensure, the

14 expectation is that full GMPs are in place for

15 manufacturing of PPQ lots to demonstrate that the

16 process is capable of consistently manufacturing safe

17 and efficacious product at commercial scale. The

18 quality controls are in place at that time, and all the

19 assays are validated. The product stability during

20 storage and shipping has been demonstrated for

21 expiration labeling.

www.transcriptionetc.com

25

1 So considering this paradigm then, where are

2 the opportunities for flexibility in manufacturing for

3 individualized therapeutics? Next slide. When

4 discussing the challenge -- thank you. When discussing

5 the challenges to the development of individualized

6 therapeutics, one must consider the complexity of

7 manufacturing biologics for a large patient population

8 and whether the challenges apply or are similar for

9 individualized therapeutics, where one would expect

10 limited manufacturing experience with an N of one lot,

11 or patient-specific lots, or small lots.

12 What then are the challenges towards achieving

13 lot-to-lot consistency and quality for the licensure of

14 individualized therapeutics? So what do we recommend

15 sponsors do to ensure manufacturing consistency during

16 standard development of a biologic? It starts with

17 ensuring quality, safety, and consistency of the

18 starting material, minimize the risk of adventitious

19 agents, and ensure reliable supply chain.

20 The manufacturing process should be optimized

21 with robust process controls and a good understanding

www.transcriptionetc.com

26

1 of the variables, which comes with manufacturing

2 experience with the release of safe, potent, and

3 quality lots that are qualified or validated early in

4 development and generating well-characterized reference

5 standards that can be used throughout clinical

6 development. Consistency in product stability can be

7 achieved by developing a platform suite of assays for

8 quality control and release testing and by using assays

9 that are qualified and validated early in development

10 and generating well-characterized reference standards

11 that can be used throughout clinical development.

12 Product stability can be achieved by adopting standard

13 or previously tested formulations, standard container

14 closures, and storage conditions.

15 What are the considerations then to facilitate

16 the development of individualized therapeutics? Go to

17 the next. In this session, as we discuss and think

18 about the manufacturing of individualized therapeutics

19 and the related regulatory challenges, it is important

20 to note that regulatory flexibility is currently

21 afforded to patient-specific biologics and biologics

www.transcriptionetc.com

27

1 that are developed for rare and orphan diseases. And

2 such flexibility is afforded based on the risk/benefit

3 analysis on a case-by-case basis. For example, in

4 early-phase studies, assays that measure product

5 potency or processed impurities and release testing

6 plan for clinical lots may not be sufficiently

7 developed or qualified.

8 There is limited product characterization, and

9 we allow products that may have limited shelf life to

10 be released, based on rapid sterility testing and

11 sampling flexibility of small lots. In early-phase

12 studies, stability testing may be limited. Product

13 stability assessments are made on data collected in

14 real time from clinical lots that are placed on

15 stability.

16 This slide lists the examples of the

17 regulatory flexibility afforded in late-phase studies,

18 which includes release of clinical lots with potency

19 assay that measures the biological activity of product

20 that may not be completely -- an assay that is not

21 completely developed or qualified. We may allow for

www.transcriptionetc.com

28

1 release of lots with wide acceptance criteria for

2 release testing of some critical quality attributes of

3 the product. When limited lots are made during

4 clinical development, we have allowed comparability to

5 be established with limited data, for example, by a

6 side-by-side analysis of critical quality attributes

7 and without robust statistical analysis.

8 Process validation in support of licensure for

9 products that have limited demand, such as orphan

10 drugs, we have allowed PPQ protocols, which is process

11 performance qualification protocols that are developed

12 for process validation, and to release for process a

13 PPQ batch for the distribution before complete

14 execution of the process validation. So we call that

15 concurrent release. And that is consistent with the

16 FDA's guidance on process validation. It is key to

17 note that, even in such cases, any lot release

18 concurrently must comply with all CGMPs, regulatory

19 approval requirements, and PPQ protocol lot release

20 criteria.

21 So with that introduction to the session and

www.transcriptionetc.com

29

1 the manufacturing issues in general, we hope that the

2 speakers and the discussion following the presentation

3 will address the challenges of small-scale batch

4 manufacturing that may be needed for individualized

5 therapeutics, ways in which we can ensure manufacturing

6 consistency for such patient-specific therapies, the

7 regulatory challenges in manufacturing and how these

8 can be addressed, and whether there is a need for

9 additional guidance, standards, or other regulatory

10 tools. So with that introduction to the session, it is

11 my pleasure to invite Dr. Guangping Gao, who is the co-

12 director of the Li Weibo Institute for Rare Disease

13 Research. He's also the director of the Horae Gene

14 Therapy Center and Viral Vector Core.

15 He holds multiple appointments at the

16 University of Massachusetts Medical School and is an

17 elected fellow for the U.S. National Academy of

18 Inventors and the American Academy of .

19 He's also the president of the American Society of Gene

20 and Cell Therapy. He's an internationally recognized

21 gene therapy researcher. He's been instrumental in the

www.transcriptionetc.com

30

1 discovery and characterization of many AAV serotypes

2 that have revitalized the gene therapy field. And he

3 has made several impactful contributions in this field

4 of AAV vector gene therapy.

5 With that introduction, I welcome you to give

6 the first talk.

7 CHALLENGES AND OPPORTUNITIES IN DEVELOPMENT AND

8 MANUFACTURING OF INDIVIDUALIZED THERAPEUTICS WITH AAV

9 VECTOR-BASED GENE THERAPIES - DR. GUANGPING GAO

10

11 DR. GAO: Thank you to FDA and our colleagues

12 here inviting me to talk about AAV gene therapy for

13 individualized therapeutics. So what I would like to

14 use this time to tell you about some basic concepts and

15 challenges in AAV gene therapy for individualized

16 medicine. This is my disclosure.

17 And so gene therapy strategy, I would like

18 first to take a broad view with you what are the gene

19 therapy approach strategies. The first one is

20 traditional or conventional medicine developments, that

21 is you use gene therapy such as AAV based vector as a

www.transcriptionetc.com

31

1 drug directly given to human patients and accomplished

2 therapeutic efficacy. And here, we have three drugs

3 approved by EMA and by FDA.

4 The second strategy is, basically, genetically

5 modify the cell therapy. In this case, you take out

6 human cells and genetically modify in vitro, expand it

7 and give back as a live medicine, a living medicine,

8 and give back to patients. Okay. And there are three

9 drugs approved by EMA and FDA.

10 So to accomplish gene therapy, I personally

11 believe there are four critical components: therapeutic

12 gene and the pathomechanisms, vector delivery methods,

13 and animal model to perform preclinical product

14 developments. And among those, the delivery vehicle

15 vector is most important. So if you look at the field

16 of the gene therapy in the past two-and-a-half decades,

17 you can see that, basically, gene therapy has been

18 going through different vector platform, includes

19 adenovirus and lentivirus.

20 So we, as the gene therapists, our wish list

21 is the following. We want the vector to have a high

www.transcriptionetc.com

32

1 efficiency, long-term stability, low immunogenicity and

2 toxicity, and no genotoxicity. So among those

3 features, adeno-associated virus has it all. So this

4 virus is really a teamwork inside-out, with the capsid

5 determining where the vector should go and how to get

6 there, where to drop the gene payload in the right zip

7 code.

8 And the vector genome itself, it actually

9 carries gene payload for gene therapy. One of the

10 major limitations that we have here is adeno-associated

11 virus is very small. It only can carry the transgene

12 cassette, no more than 4.6, 4.7 kb.

13 And however, this genome is critical in terms

14 of formed, stable episome vector structures to be

15 persistent in terminally-differentiated tissues. So

16 how we transform this almost a virus -- because without

17 helper, this virus is doing nothing -- into amazing

18 vector. So basically, what we do is we take out wild-

19 type genome section of regulatory protein and a capsid

20 protein. And then we put back our expression cassette,

21 therapeutic gene. And then we provide helper function

www.transcriptionetc.com

33

1 in trans. We provide the packaging plasmid, the

2 regulatory sequence, and capsid sequence in trans.

3 If we put those three things together, either

4 by transient transfection, stable transfection, or

5 infection, we can create -- get into the producer cells

6 and then generate AAV vectors. The beauty of this

7 virus is that what AAV dressed doesn't matter. So what

8 you can do simply is change the dress, change the

9 capsid and become a new vector. For example, this we

10 change to AAV8. 9 capsid become AAV9 vector. And then

11 if you change to AAV8, it becomes AAV8 vector.

12 So exactly how AAV works, I just want to use a

13 simple cartoon to demonstrate how AAV works as a gene

14 therapy drug. Basically, like any other viruses,

15 AAV’s getting to the cells by receptorology process,

16 and then going through endosome trafficking. There you

17 have two pathways. One, you probably can get into a

18 proteasome, get degraded. And another, you can enter

19 the nucleus. And efficiency or the ratio of those two

20 proportions of the viruses depends on serotypes.

21 Some serotypes have more efficient escape from

www.transcriptionetc.com

34

1 endosome, and some it's more efficient to get into the

2 cells, into the nucleus. Once you get into the

3 nucleus, the first step AAV need to uncoating and then

4 that release, let single strand AAV genome are both

5 negative strand and positive strand. Then you can form

6 double-strand genome through host machinery or self-

7 annealing. And then, you generate a vector genome that

8 is a ready to expression.

9 However, there's a mechanism because of a

10 terminal repeat of AAV8. It can form a circular

11 version and then can be stabilized in a terminally-

12 differentiated tissue. Very small portion of AAV could

13 be potentially integrated.

14 And then once you have a stabilized genome

15 that you have a gene expression, you can generate gene

16 therapy products in this case. So if you look at the

17 AAV gene therapy -- and currently, AAV gene therapy has

18 entered an exponential growth period. And there are

19 nine different therapeutic areas using AAV gene therapy

20 vector.

21 Ophthalmology or ocular gene therapy and CNS

www.transcriptionetc.com

35

1 gene therapy takes the lead but followed by many other

2 different applications. So now we know -- we talk

3 about this strategy. We talk about the vector

4 delivery. And exactly how can you accomplish gene-

5 based therapy, I summarize here in four different

6 formats.

7 The first one is a classic gene therapy, that

8 when you have a gene not functional, you can provide a

9 healthier gene to replace these malfunctioning .

10 This is a very classic version of the gene therapy.

11 The second part is that you may have nothing wrong with

12 your gene, but you need certain gene products to fight

13 diseases such as cancer or infectious diseases. You

14 can add genes to fight those diseases.

15 The third one is, when you have a genetic

16 , when you get a gain of toxicity, gain of

17 function outcome, you can silence this gene and

18 accomplish therapeutic treatment. And the final one

19 could be gene editing. And this is now entering a

20 rapid growth using gene editing as a tool for gene

21 therapy.

www.transcriptionetc.com

36

1 So now, I would like to use one of my

2 personal-professional journey about a disease called

3 the demonstrating how gene therapy

4 works. This disease was discovered in 1931 by Dr.

5 Canavan. And this is a very tragic disease,

6 devastating disease.

7 Basically, you have a quite high prevalence in

8 Ashkenazi Jewish population, but also, in general

9 population, you have 1 in 300 . The genes

10 could've been mutated. And then this patient dies

11 usually before five. There's no medicine to treat this

12 patient -- those patients. And this is a spongy

13 degeneration, the disease is.

14 And this is a picture I took in 2013, when I

15 went to a Canavan disease meeting and I took this with

16 a dozen of the patients. And 2016, I went back to the

17 same meeting, only a few kids still running around.

18 And this really picture remind me, we as gene therapy

19 researchers, we have to work as hard as we can to have

20 the race with time to develop a therapy to save the

21 lives of those children.

www.transcriptionetc.com

37

1 And as you can see, because of this

2 is a spongy degeneration, the ,

3 whole brain is just like Swiss cheese. And this is my

4 mentor, PhD mentor, Dr. Rueben Matalon, who discovered

5 biochemical defect of this disease in 1988. Basically,

6 what happened, there's a product called NAA made in

7 , in mitochondria. And they transport into

8 where it should be degraded by

9 aspartoacylase.

10 However, if we have this gene mutation, then

11 all the tragic starts here. You have NAA accumulating

12 in inter-tissue space in the brain, and then you

13 generate spongy degeneration. I entered this Canavan

14 research in 1989 as a graduate student and published my

15 thesis research in 1993. And this allows the gene

16 therapy possible. But the issue is, as everybody knows

17 that -- the key issue, as I said from the beginning,

18 it's the vector.

19 So I decide to start my post-training with Dr.

20 Jim Wilson at the University of Pennsylvania. And

21 then, Jim and I started this joint venture that is

www.transcriptionetc.com

38

1 looking for more efficient AAV. And we basically

2 designed primers across the variable regions of the AAV

3 genome, amplifying the capsid. Using this method, we

4 generated a library of AAV capsids.

5 And I want to give you an example. This is

6 one of the most expensive but most popular virus,

7 that's AAV9 -- everybody knows that, AAV9, so widely

8 used for different applications in gene therapy. So we

9 isolated this from human tissue, human liver. And

10 that's the original lab record showing what isolates

11 clone 28.4 as a gene AAV9 vector. And the beauty of

12 this vector is it can cross blood/brain barrier and

13 transduce the brain quite efficient.

14 So with this available, then we, at the

15 University of Massachusetts, start to develop gene

16 therapy for the disease. This is my MD PhD fellow, Dr.

17 Dominic Gessler. So basically, we developed it using

18 AAV9. And through extensive optimization, we developed

19 the vector treat an animal model exactly recalculate

20 human disease.

21 As you can see, the gene therapy here,

www.transcriptionetc.com

39

1 basically, this is Canavan disease. The neurotrack,

2 it's very short. This is normal. But after gene

3 therapy, you basically, through this tractography, you

4 see we restore neurotrack in the patient mice.

5 And very importantly, this gene therapy can

6 reverse pathology. As shown in here after one week of

7 gene therapy and this Canavan disease still there. And

8 this is wild-type, and this is Canavan treated there,

9 so not yet. But if you look at week four, then the

10 situation will be very different. And this is the

11 Canavan disease that we have treatment, this is normal,

12 this is treated. As you can see, we basically restored

13 myelination and reversed pathology.

14 So with this available -- and I collaborated

15 with Dr. Barry Byrne of the University of Florida -- we

16 started as a Phase 1 -- not Phase 1, sorry -- expanded

17 access trial with two-year-old patients. And

18 basically, we -- in this process, we also applied

19 immunosuppression to leave a potential window of

20 treatment for future dosing if this treatment didn’t

21 work. And as you can see, treatment is safe.

www.transcriptionetc.com

40

1 Transaminitis, we do not have transaminitis elevation.

2 And also, this patient growth curve, as showing here,

3 is quite encouraging.

4 And so if you summarize the data from this

5 expanded access, is we know the immune modulation

6 worked perfectly because up to today, we still cannot

7 detect any anti-AAV9 antibody in the patient's

8 bloodstream.

9 And also, AAV gene therapy improved the

10 myelination and also the motor development and restored

11 the vision. The patient could not see before. Now,

12 when you walk in the room, the patient can follow you

13 around with his eyes.

14 And also, using DTI, the MRI, we demonstrate

15 that we show the development findings as we saw the

16 patients start moving with their hands. And this is a

17 patient before the treatment, and this is the patient

18 about, I would say, 15 months after treatment. You can

19 see, really, we improved their mobility and also

20 quality of life.

21 And whenever I saw these pictures, I feel very

www.transcriptionetc.com

41

1 much rewarding as a scientist who spends your life and

2 your professional research on this disease. The family

3 will be able to see the patient can save his life and

4 improve the quality of his life. So this is a powerful

5 -- a powerful outcome of the gene therapy. However, I

6 want to remind you, with gene therapy, we still have a

7 long way to go.

8 And in this journey of gene therapy express

9 reach to hospital usage and clinical application, we

10 have several hurdles we're going to face. The first

11 one, number one, I can see is the manufacturing. And

12 we -- I don't think we have enough vector to treat

13 patients as a commercial drug. And particularly, the

14 potency of the vector, we also need to improve.

15 The second part is this pre-existing and also

16 adaptive capsid transgene responses. And this can --

17 immunotoxicity could generate safety concerns. The

18 third part is, even though we want to say we should

19 have cell-specific, tissue-specific gene therapy -- but

20 I have to tell you, at this stage, my personal belief

21 is we only have efficient vector and nonefficient

www.transcriptionetc.com

42

1 vector. I don't think we really can control tissue

2 tropism and cell specificity.

3 And the final hurdle is, so far, we thought

4 the gene therapy, the more the better. Actually, soon,

5 I personally believe we will see that we need to have a

6 regulated gene therapy, long-time, overexpressed, super

7 physiological level may not be good. So once we have

8 those hurdles overcome, we'll have regulated gene

9 therapy arrow, have as a drug for clinical usage.

10 So how to address those questions? I want to

11 give you one example. That is we continue looking for

12 better vectors and for -- basically, this is an effort

13 we have done -- and generate about -- so through the

14 PCR and the high-throughput sequencing, we generate

15 about 70,000 more AAV capsid. Through complicated

16 bioinformatics pipeline, we eventually identify about

17 1,000 vectors. And some of those vectors can overcome

18 the production hurdle. AAV2 is one of the poorest

19 producers, as you guys doing gene therapy should know.

20 But we found in those variants, AAV2, 25

21 percent of them produce much better than AAV2. Some of

www.transcriptionetc.com

43

1 them produce 20-fold higher than AA2. And I want to

2 show you one example of such efforts. And that is we

3 isolated AAV2 variants, only have 13

4 difference from AAV2.

5 However, this translates to -- into 13-fold

6 more efficient than gene delivery, seeing as you can

7 see here, you have AAV2 injected in one side of

8 hippocampus. And then if you look at other side

9 injected with new vectors, you can see 13-fold of

10 enhancement. It tells you we still have the room to

11 improve our vector.

12 And we also continue this kind of search. The

13 first one, we screened about 38 vector, and then here,

14 we screened about 50 vector or so. We identified 21

15 vector that's as efficient as AAV9. Seven of them, I

16 actually can see either equal or pass AAV9 performance.

17 So it tells you we have further room to improve

18 identify more efficient vectors for CNS gene therapy.

19 So -- but now, for gene therapy manufacturing,

20 that's one of the topics of today's meeting. And so

21 far, the current platform technology, there are two

www.transcriptionetc.com

44

1 categories.

2 One is transfection-based. This is developed

3 by Drs. Xiao Xiao and Jude Samulski in 1998. This is a

4 very popular process for AAV manufacturing. And then

5 if you look at the other category that's infection

6 based, either HeLa -- producer cell line HeLa, rep/cap

7 cell line, herpes virus, or Sf9 Baculovirus. Those are

8 infection-based process.

9 But if you look at the current -- I give a

10 snapshot of where we are in terms of manufacturing --

11 you can see here 293 transfections still dominate the

12 manufacturing process. And second is a Baclovirus

13 quickly catch up. And third on is a HeLa-based -- the

14 herpes virus or adenovirus-based and herpes virus-

15 based. So this is kind of current trend in

16 manufacturing platform technology. However, if you

17 look at the whole field, you will see CDMO's play a

18 major role, only about 30 percent of the company, doing

19 AAV themselves, and CDMO becomes a rapid-growth

20 industry here.

21 So now, for AAV manufacturing, what are the

www.transcriptionetc.com

45

1 major challenges we are facing? So I would say it

2 depends on many different factors, such as targeted

3 tissues, eyes, versus muscle, and route of

4 administration, subretinal injection versus systemic

5 injection and patient population size or patient size,

6 such as pediatric versus adult patients, and the

7 serotype and transgene cassette also contributed to

8 manufacturing. But the gap between the current

9 producibility and the clinical needs is either a one-

10 log, two-log, or three-logs, depends how big the

11 patient population size is.

12 So the second issue is how are we going to

13 close this gap? I personally believe we need -- really

14 cannot just do by increasing the bioreactor size, and

15 we need to enhance yield per cell. This will

16 definitely require collaborations between vector

17 biologists and bioengineers.

18 So one thing we should consider is AAV

19 replication and packaging biology, understand there's a

20 timing sequencing and a level of different gene

21 expression. The second part is cellular factor. We

www.transcriptionetc.com

46

1 have to figure out the cellular factor that can further

2 boost replication of packaging. And third is, ideally,

3 we should have a producer cell line, real producer cell

4 line, just adding the compounds that trigger entire AAV

5 manufacturing.

6 And this we have a lot of work to do ahead.

7 Of course, like any other FDA-approved product, quality

8 control and the bioanalytics is very important. One of

9 the major issues with AAV is empty-to-full ratios. And

10 right now, because it's manufacturing we’ve still not

11 optimized yet, it’s 90 percent empty to 10 percent

12 full, and if you do not have a special process to

13 purify it -- but our goal is to reverse it, 10 percent

14 empty and 90 percent full.

15 The second part is a biopotency assay,

16 infectivity assay. This is very hard because, ideally,

17 you should develop a kind of across different product

18 pipeline, infective assay. And we also need a

19 biopotency assay. This is product dependent.

20 And finally, it's about the vector genome

21 itself. And currently, for many different reasons, we

www.transcriptionetc.com

47

1 really do not understand what we have in AAV package.

2 I'm going to show you an example here. So if you put

3 guide RNA into this construct, if there's a simple

4 guide RNA structure, you can see you get a uniform

5 band, single band. However, what's exactly in this

6 band, we do not know.

7 My colleague, Phil Tai start looking to this

8 black box of AAV. He developed this sequencing for ITR

9 to ITR, every single molecule of AAV prep and to

10 understand exactly what's there. You can see the

11 majority of the vector here is full-length.

12 And however, if you put two guide RNA into the

13 vector, and then in these head-to-tail -- head-to-head

14 configurations, you can see you have many truncated

15 genomes. And if you use the sequencing we call the

16 AAV-Gp Seq we consider as a next-generation QC

17 pipeline, you can see you virtually have no full-length

18 molecule. You have many different truncated forms of

19 the molecule. If without this technology, you do not

20 know exactly what you have.

21 And however, interestingly, if you change that

www.transcriptionetc.com

48

1 configuration to head-to-tail of the two guide RNA,

2 then you come predominantly a single band. And this is

3 confirmed by sequencing itself. So this tells us we

4 still have a way -- a lot of work to do to figure out

5 quality control and give the best medicine, safest

6 medicine to our patients.

7 And so finally, I want to summarize what's the

8 difference between this individualized therapy-based

9 traditional gene therapy. And so we have one problem.

10 We have realized that, pre-clinical development, we

11 need better understanding the genetic causes and the

12 pathomechanism.

13 Depends on -- it’s loss of function, gain of

14 function, autosomal recessive versus dominant, X-link

15 or haploid insufficiency, this -- you will have a

16 different strategy for gene therapy. And also, many

17 ultra-rare diseases, there's no animal model for it.

18 So this in vitro versus in vivo model versus normal

19 versus disease model, expressing versus the function

20 and versus the phenotype correction, those are the

21 things we have to figure out at least, those ultra-rare

www.transcriptionetc.com

49

1 diseases with the patient of 1 to 10 to 100.

2 And also, the timing and the cost remain the

3 same. If you do a gene therapy drug to preclinically

4 develop this drug, it's the same cost, the same time.

5 It doesn't matter it's 10 patients or 10,000 patients.

6 And I don't think from NIH funding point of view, when

7 you -- disease from a small -- I don't know how much

8 funding opportunities we could get.

9 And the regulatory and supportive flexibility,

10 again, to generate this data to support in vivo data

11 for efficacy to help prepare IND and prepare the

12 pivotal toxicology studies, I think the time and

13 regulatory support required is same. Finally, GMP

14 manufacturing and the QC bioanalytics, so most of the

15 challenge remains the same. It doesn’t matter this

16 drug for 1 patient, 10 patients, or 10,000 patients.

17 And additional unique challenges include technology

18 transfer and the process development lot cost remain

19 the same. It doesn't matter it's 1 lot, 1,000

20 patients, or 10 patients.

21 And cost for the smaller batches may not be

www.transcriptionetc.com

50

1 even linearly scaled down. You probably cost more for

2 small-pack batches. And 1 dose versus 100 doses, if we

3 have existing patient treated with 10 patients treated,

4 what about a future patient? Where we should store and

5 maintain the stability and distribution of those gene

6 therapy vectors? I think as that's some of those

7 topics we should discuss after this meeting. Thank you

8 very much.

9 DR. TARAPERWALA: All right. Thank you, Dr.

10 Gao. Our next speaker is going to be Dr. Jason Gill

11 from Texas A&M. He was born and raised in Canada and

12 received his BSc and MSc degrees from Brock University

13 and his PhD in Food Microbiology from the University of

14 Guelph in Canada.

15 He holds a faculty position at the Department

16 of Animal Sciences at Texas A&M University, where the

17 major research focus in his lab is the biology and

18 application of bacteriophages. These are often also

19 referred to as phages. And you're going to hear more

20 about it. These are viruses that can infect bacteria.

21 Specifically, the research in his lab includes

www.transcriptionetc.com

51

1 phage genomics, basic phage biology, and application of

2 phages in real-world settings against many bacterial

3 pathogens. Dr. Gill holds many joint appointments in

4 the faculty of genetics in the Department of Microbial

5 Pathogenesis and Immunology in the College of Medicine.

6 And he serves as the associate director for the Center

7 for Phage Technology and Interdisciplinary Research and

8 Teaching Initiative, supported by Texas A&M AgriLife

9 Research. So welcome, Dr. Jason Gill.

10 DEVELOPMENT OF PHAGE THERAPY: PERSONALIZED MEDICINE AND

11 INDIVIDUALIZED THERAPEUTICS - DR. JASON J. GILL

12 DR. GILL: So good morning. I'd like to thank

13 the organizers for inviting me here to give this talk.

14 So what I'm going to talk about -- what I was asked to

15 talk about development of phages, as a therapeutic and

16 really, more on the manufacturing side. And so to

17 understand the manufacturing part, we're going to have

18 to understand more about the biology of that as well.

19 This is my COI statement. So I did some consulting for

20 Merck last year.

21 So to understand about the development of

www.transcriptionetc.com

52

1 phages as a therapeutic, we have to understand that

2 phages are very diverse. And so Dr. Gao talked about

3 adenoviral vectors. And they're quite variable. And

4 so phages are probably the most diverse set of

5 organisms on earth. They're very ancient, and they've

6 been coevolving with bacteria for many billions of

7 years.

8 They tend to be very adaptive to their hosts

9 at the strain and species level. So they're very

10 specific. They have very diverse genome content. So

11 you'd have two phages that infect the same host which

12 have really zero detectable DNA sequence similarity at

13 all. So there's a large diversity there in terms of --

14 that drive how you do your developments. And there's

15 also a lot we don’t understand about how phages work on

16 a more fundamental level.

17 So a lot of phages carry like hypothetical

18 genes, genes of unknown function, which as far as we

19 can tell have no phenotype. But again, for regulatory

20 purposes, that may become more important. So the

21 development of phages, this is actually not a new idea.

www.transcriptionetc.com

53

1 So phages were actually used as a relatively mainstream

2 therapeutic up until the 1940s when antibiotics were

3 developed. And then they were kind of abandoned in the

4 west and revived really with the rise in antimicrobial

5 resistance, starting really intensely about 20 years

6 ago.

7 So this is from an older paper. But this lays

8 out two theoretical development pipelines for using

9 phages. So the top one, you have this -- what's called

10 “pret a porter” system. And this is what was talked

11 about in the opening talk with this idea that you kind

12 of have a mass-produced predefined product. So you'd

13 isolate a phage or a set of phages against a particular

14 pathogen. You would understand them as well as you

15 could. You would have a GMP manufacturing process.

16 You take them through a regular phase one,

17 two, three approval pipeline. And then, you would mass

18 produce that product and then market it. You'd have

19 like a CGMP-produced, very well-defined product.

20 And the alternate approach on the lower panel,

21 in blue, is this kind of “sur mesure” idea, which is

www.transcriptionetc.com

54

1 this made to order idea. And so this is that you would

2 have a collection of phages, maybe 100 phages, maybe

3 200 phages, 500 phages, in a library. You would get

4 bacteriology back from a patient.

5 You would then identify phages that worked

6 against that particular -- that patient's particular

7 strain because remember phages are very specific for

8 their hosts. And you would then work that up into some

9 kind of product then administer that to the patient.

10 So the idea there is that when you have this

11 kind of product, you have tying to the bacteriology.

12 But you have a lot of issues on the regulatory side.

13 So the product identity is not as clear in that kind of

14 situation. The manufacturing is likely to be less

15 controlled. And this really thought of -- this is

16 about a 10-year-old paper. And this was thought of as

17 really kind of a -- not necessarily with the idea of

18 developing a large commercial product in mind. This is

19 really a way to treat patients.

20 And so this kind of approach works well if you

21 want to just kind of keep doing like expanded access or

www.transcriptionetc.com

55

1 eIND administrations, which have been happening. But

2 it's hard to see how that would be commercializable.

3 So I think what we're talking about here today is this

4 personalized medicine approach, where you would want to

5 take part of that sur mesure idea, where you have this

6 library of phages, and try to then take that through

7 some kind of regulatory process. So that could

8 actually be a widely available, possibly even mass-

9 produced product.

10 So product identity is pretty important for

11 any kind of therapeutic. And so on the -- if you have

12 a fixed product, that's a lot easier to define. So

13 you'll have a phage isolate or a set of phage isolates.

14 They'll be grown on a host or a set of hosts. There

15 will be some kind of production and purification

16 process that will be defined. And then you end up with

17 a product at the end. So that's a lot more straight-

18 forward, even for a biological.

19 And personalized approaches becomes -- the

20 idea of product identity becomes a little fuzzier. So

21 if you have, say, a collection of phage isolates, they

www.transcriptionetc.com

56

1 may or may not be grown on the same host. So you're

2 going to have that variance. You're going to have not

3 just new phages but phage host pairs in the

4 manufacturing process. There's going to be possibly

5 different manufacturing processes for different phages.

6 They may not all work in exactly the same manufacturing

7 process.

8 And then you're going to end up with,

9 hopefully, some CGMP manufactured individual phage

10 isolates, which you then blend together and then give

11 to a patient. And then so when you're talking about

12 product identity -- so is the product identity each of

13 the individual phages? Is it the final mixture of

14 phages? And right now, I don't know how -- if that

15 aspect is very clear.

16 So the size of the library you're going to

17 need is also going to be pretty variable between things

18 you want to treat. So different pathogens have

19 different levels of diversity, and that will affect the

20 size of the library that you're going to need to have.

21 So if you have very diverse targets, like for example,

www.transcriptionetc.com

57

1 Acinetobacter baumannii or Burkholderia cenocepacia,

2 which are organisms that we deal with, the host range

3 of those phages is really narrow.

4 So on the right, there's some data from our

5 lab on some collections of phages against Acinetobacter

6 baumannii. And this isn't the full table; this is just

7 the table that shows where the hits are. There's

8 another panel about this size, where the phages also

9 don't infect any of those strains. So the host strains

10 with these phages are very narrow. But if you look at

11 organisms like, for example, staph aureus or listeria

12 monocytogenes or shigella, those organisms are much

13 more clonal. And in that case, you can have a smaller

14 phage collection, which will cover most or more of

15 those strains.

16 So the target really affects the diversity of

17 the library. So if you're talking about a personalized

18 approach, if you're talking about this for staph

19 aureus, you may need a library of only 20 phages or 10

20 phages. But if you want to do this for Acinetobacter

21 baumannii, you're probably going to need a few hundred

www.transcriptionetc.com

58

1 on hand if you want to actually be able to cover most

2 strains of the pathogen.

3 So phages can be developed in a few different

4 ways. So they can be completely natural. They can

5 contain engineered components. This is -- some of this

6 also is still really evolving in the field. So you can

7 have the engineering be present in the phage

8 . Actually, when the phage replicates, that

9 is a transmissible change, or they could also be

10 expressed in trans. So really, just the phages you

11 produced have the engineered or modified component, but

12 they don't pass that on to their offspring. Or they

13 may not produce any offspring at all, depending on how

14 you've engineered them.

15 There's a few companies that are looking at

16 synthetic phages right now. But you have to keep in

17 mind, even when they're talking about a synthetic

18 phage, it's still based on natural DNA sequence from

19 other phages. It's just been pieced together. So

20 there's a few different ways then that these are being

21 developed. There's a replicative phage therapy, which

www.transcriptionetc.com

59

1 I call this the classic approach.

2 So this is you're taking natural phages from

3 the environment, selecting them, working them up into

4 some kind of treatment, and then they are going to

5 infect cells. And they're going to replicate and lyse

6 those cells and release progeny. And that can happen

7 with naturally occurring phages or with engineered

8 phages.

9 And another route which is being explored is

10 using highly engineered phages, which don't necessarily

11 replicate. And they will -- they might just adsorb to

12 a cell and then deliver some kind of cytolethal agent,

13 like a CRISPR protein or something else that would

14 cause the cell to die. Or it's possible to have a

15 phage deliver a genetic payload to a cell to alter its

16 phenotype, for example, to just remove resistance

17 plasmid. Or it's even been proposed to say allow the

18 cell to now start synthesizing new metabolites and that

19 -- this is like a microbiome engineering type of thing.

20 But these are various ways that you can --

21 they are neat phage tricks that you can do. So they're

www.transcriptionetc.com

60

1 quite versatile in how you can engineer them. And this

2 isn't exhaustive either. I'm sure people are thinking

3 of all kinds of wild new ways you can use phages.

4 So one of the limitations here is the

5 knowledge base of phage biology as it exists today. So

6 phages are relatively complex viruses. This is a map

7 of the genome of phage lambda. And you can argue that

8 phage lambda is the best understood organism in biology

9 because of its long history of study. It has about a

10 49-and-a-half kb genome, as you can see here. But even

11 though this phage has been studied for, like, thousands

12 of person years of work likely, there are still some

13 genes we actually -- the functions are not known.

14 So if you can delete those genes, they don't

15 appear to have a phenotype. And those are highlighted

16 there in green. The rest of it is all pretty well

17 understood.

18 But then we move that focus from lambda. This

19 is what we call an inch wide and a mile deep. So we

20 know a lot about lambda. If you move that focus over

21 just a little bit, this is a group of phages that are

www.transcriptionetc.com

61

1 morphologically similar to lambda.

2 They infect salmonella. They're called 9NA-

3 like. 9NA is the first of this type to be isolated, so

4 that's what they're all called. So they have a

5 relationship to lambda. They're roughly the same size,

6 a little over 50kb. But if we look at this group of

7 phages, there's 176 genes in this group of phages.

8 Only 32 of those genes actually have a

9 predicted function. And 143 don't have a known

10 function, at least not what we can tell

11 bioinformatically. And 68 don't even have a

12 counterpart in the database. So they're completely

13 novel genes.

14 And this is not that unusual when you're

15 finding new phages against new bacterial targets that

16 you'll have a section of protein-coating genes.

17 Actually, they're the first example of those genes that

18 have been found in biology. So we've done this for

19 Caulobacter. And in that case, I think something like

20 two-thirds of the genes were actually novel in those

21 organisms.

www.transcriptionetc.com

62

1 So phage production is going to be based

2 around -- it's a biological manufacturing process. So

3 they're generally going to have to be propagated in a

4 live host. It's not very hard to get titers of 10 to

5 the 9 or 10 to the 10 pfu per ml in a culture. That's

6 just in the lab. If you optimize that, you can

7 probably drive that up to 10 to the 11 or maybe 10 to

8 the 12 even.

9 There's a few different ways you can get from

10 your crude production lysate down to the final product,

11 which are up here. So you're starting with a lysate to

12 somehow -- and you can use a few different methods to

13 take you from that, which is going to be the crude

14 liquor, basically, of the culture, down to somewhat of

15 a clarified lysate. And then there's a few

16 technologies that exist to take you from that to kind

17 of a relatively pure concentrate, which you can then

18 put through additional polishing steps, too, so we can

19 help remove endotoxin or further purify it.

20 So the clarified lysate is really just going

21 to be spent cultured media with phage in it. It's

www.transcriptionetc.com

63

1 going to have a lot of cell debris and media components

2 in it. A clean concentrate might be relatively pure.

3 It's mostly phage. And it will be more concentrated.

4 Most of the media components have been removed

5 and the cell debris. The endotoxin level will be

6 reduced. It may not be as low as you need, so you may

7 need to do additional steps to remove endotoxin or to

8 remove additional contaminants until you get to a

9 purified phage, which you would then blend and package.

10 So the methods are still very much in

11 development. And the different companies have their

12 own internal processes for that. But they're not

13 completely alien, I don't think, to the world of

14 manufacturing. This is a -- they're not that

15 different, say, from production of common proteins from

16 bacteria or certain viral vectors or vaccines. You're

17 having a biological production process. The

18 technologies aren't -- they're comparable; they're not

19 completely different.

20 But also, it's important to remember that the

21 production process here is likely to be a major part of

www.transcriptionetc.com

64

1 a company's IP around that product because the phages

2 you can't necessarily patent themselves if they're

3 natural. So one of the things you can have you can

4 patent, and also even protect the trade secret, is the

5 manufacturing process. And so you have to keep that in

6 mind when it's going through the regulatory process.

7 So the purification method that you use will

8 probably then determine that product identity. And

9 also, you have to remember that, because phages are

10 very diverse, not all phages are going to necessarily

11 respond the same way to the same manufacturing process.

12 So you may have to have, basically, a -- you might have

13 a set process that you're going to have to have

14 variances built in for or maybe even several different

15 parallel processes, depending on the phage you want to

16 produce. And they can vary in terms of their pH

17 stability and their surface charge, their stability in

18 various buffers, and formulation agents. And so you're

19 probably going to need to be able to have room for

20 adjustment depending on different phages from that --

21 from a library.

www.transcriptionetc.com

65

1 So just a note about phage production though.

2 So the technology really to maximize that yield would

3 really be a big help in this field. So this is a

4 picture from the Patterson case that we were involved

5 with, with the groups from UCSD -- and Dr. Schooley’s

6 here -- and the group from the Navy.

7 And so when phages were actually administered

8 to Dr. Patterson, they were administered as doses of 10

9 to the 9 pfu and diluted into 100 ml Ringers and

10 infused. So 10 to 9 pfu is actually not that much. We

11 can actually grow cultures of 10 to the 9 to 10 to the

12 10 pfu per ml in the lab. And you can grow up a couple

13 liters of that even just in the lab scale.

14 And so in theory, one liter of culture could

15 produce like a thousand doses of this phage for a

16 patient. But in practice, by the time we actually went

17 through our purification process, our recovery was

18 something like 0.1 percent. So we had to produce

19 multiple batches. So any technology that you can use

20 to -- if you can recover even half of the initial

21 particles that are in the -- that initial culture, that

www.transcriptionetc.com

66

1 makes it much more efficient than at least what we've

2 been able to do in the lab so far in a rush. So having

3 an optimized production process certainly helps.

4 So if we're looking at then producing phages

5 possibly of different strains, so the variation of the

6 host is also going to be a contributing factor to your

7 product identity. And it's going to determine partly

8 the contaminant profile. So if you had a very diverse

9 phage collection, you're likely going to have more than

10 one host you're going to have to grow all these guys

11 on. And so different strains of the same species, they

12 can vary in their toxin production, their LPS structure

13 and how pyrogenic it is. Mobile element content will

14 change. And also, many bacterial strains carry

15 multiple prophages.

16 And this is just a map from a relatively old

17 review. But this shows maps of -- these are actually

18 active prophages in bacterial genomes. And so it's not

19 that unusual for the average bacterial genome to carry

20 one to three functional prophages. And when you

21 propagate the phage, your phage, your virulent phage,

www.transcriptionetc.com

67

1 on that strain, some of that prophage will be induced

2 and will end up in that culture as well.

3 It likely is a very minority component. For

4 example, if you have 10 to the 10 of the phage you

5 want, you might have a 10 to the 4 or 10 to the 5 per

6 ml of this other phage. But it's going to be there,

7 and it's going to vary depending on the strains that

8 you want to use.

9 And these are an issue because they could

10 possibly, in theory, transduce DNA from your

11 propagation host into another host in the patient or

12 possibly between strains in the same -- in the patient

13 once they're administered. Maybe, right? These are

14 all just theoretical possibilities.

15 It is possible to take bacterial strains and

16 engineer them to reduce their contaminant profile. So

17 this is an example of the bacillus strains, the root of

18 strain 168 that as it's called MG1M, which is developed

19 by, I believe, a Japanese group. And really, they just

20 took the bacillus strain 168, which is kind of the go-

21 to model strain for bacillus subtilis, and they removed

www.transcriptionetc.com

68

1 all the prophages and the polyketide synthesis and a

2 whole bunch of other stuff they thought the cell didn't

3 really need. And they ended up removing about 23

4 percent of the genome.

5 And this was for a -- for using the cell as a

6 biofactory. They wanted to be able to put your own

7 metabolites in here. It will grow more efficiently and

8 make more of the or whatever it is that you

9 wanted. But you could theoretically do the similar

10 thing here for phage hosts as well and remove all the

11 components that you don't want from that genome. It's

12 not a -- it's a nontrivial undertaking to do this.

13 So if you had to do this, say, with 50 strains

14 to propagate your phage, that will be -- that would be

15 an undertaking but certainly doable. It also maybe is

16 possible to generate like a universal host or at least

17 a much broader host range host that you have. So

18 instead of having to take your hundred phages and

19 propagate them on 50 different hosts, you would

20 propagate them on five different hosts, which would

21 certainly simplify your manufacturing process. It's

www.transcriptionetc.com

69

1 also maybe possible that you could express phages from

2 a completely orthologous host. For example, in yeast,

3 they could be recombinantly produced in a yeast cell or

4 some other method. Again, I'm not aware of anybody

5 actually doing that, but maybe it's possible.

6 So product identity, also the -- in terms of

7 active ingredient concentrations, are important. So in

8 a phage product, the actual concentration of active

9 ingredients is likely to be very low, in the order of a

10 microgram per ml or less. So the kind of standard HPLC

11 peak or something you're looking at for a drug is

12 likely not going to work here. And so the way people

13 normally evaluate the potency of the product then is

14 turn to an active titer. You're looking for plaque-

15 forming units on a lawn of bacteria.

16 And so that's the way we have done it. As far

17 as I know, that’s the way other groups are doing it as

18 well. And so you actually are measuring infected

19 particles, which is nice. They have to be viable to

20 form a plaque. But you also have to be aware that the

21 number of plaques you'll get can vary a lot depending

www.transcriptionetc.com

70

1 on the host that you put the strain on and the plating

2 conditions.

3 So this is an example from our group of some

4 staph aureus phages. And you can see the plating

5 efficiencies can vary by half a log, depending on the

6 host you put them on. So you can get -- if you have

7 your phage product, if you titer on one host, they'll

8 tell you you have 10 to the 9. A different host might

9 tell you you have 5 times 10 to the 9.

10 And so you have to think about these plaque-

11 forming unit assays. It's kind of that's kind of the

12 minimum number of viable phages that are in there. If

13 you could find a better host that gives you more

14 plaques, then that would kind of -- that raises that

15 floor up.

16 Another issue then is product impurities, as

17 well. So crude lysate contains media components and

18 bacterial cell debris, and it's going to have endotoxin

19 in it if it's a gram-negative phage. And these range

20 from relatively harmless, right? You're going to have

21 some sugars in there. And also, it could be very

www.transcriptionetc.com

71

1 dangerous like endotoxin. And there has to be

2 something -- a decision arrived at really on what

3 impurities you can tolerate in the final product.

4 And this is probably going to vary depending

5 on how you're administering it. So if it's going to be

6 a phage product for oral or topical use, you could

7 probably tolerate more impurities than you would for

8 something which is going to be introduced parenterally.

9 And again, this is -- in, at least where I am in the

10 field, nobody really knows what the rules are right

11 now. So that's something that has to be developed, I

12 think in conjunction with the regulators and the people

13 who are actually manufacturing this to decide what are

14 the acceptable impurity profiles that are going to be

15 tolerable because that will then also drive the

16 manufacturing process as well.

17 The other issues around phages, which are not

18 really necessarily manufacturing-related, but they're

19 intellectual property related. So patent protection

20 for phages are difficult to determine. They are

21 products of nature. So engineered phages are certainly

www.transcriptionetc.com

72

1 more patentable than the natural ones.

2 There's a lot of prior art dating back to the

3 1920s. For example, there are patents that exist that

4 they have patented the idea of using phages to treat

5 bacterial infections, an idea which was invented in, I

6 believe 1917 or 1918. So it's more difficult, then, to

7 get intellectual property protection.

8 And if your product is actually a viable

9 phage, it's going to replicate in the host, you can't

10 necessarily rely on trade secrecy either because

11 anybody can just take your products and just culture

12 the phage out of it. And then, they've got the phage.

13 So trade secrets aren't going to work there either, so

14 some kind of intellectual property protection is still

15 -- that's still evolving.

16 They're very narrow spectrum. So I think it's

17 harder to get companies to get involved in this field

18 because you're developing very narrow-spectrum

19 treatments, which are -- you're not going to be able to

20 cross-market them very broadly, so that makes, I think,

21 a lot of companies reluctant to invest.

www.transcriptionetc.com

73

1 And really, the idea around this is the

2 traditional business model of a single mass-produced

3 antibiotic aren't going to work, of course, for

4 personalized medicine. But something we've been

5 hearing a lot lately in the last few years is about

6 just how the model of antibiotic development in general

7 -- a lot of companies have just gotten away from

8 developing antibiotics because they're not profitable,

9 because they're -- you're not going to have a large

10 market. They’re expensive to develop.

11 And if that's true for small molecules, it's

12 also true for this. And so if there's a fundamental

13 issue with the economics of this developing anti-

14 infectives, that's less of a scientific issue. I think

15 it's more of an economic or a policy issue. But it

16 also -- that's going to drive the availability of these

17 products.

18 So just to wrap up, so some solutions that --

19 to address some of these things, we need better

20 understanding of some of the non-paradigm phages that

21 are infecting different hosts. Like we know a lot

www.transcriptionetc.com

74

1 about a few phages that infect e coli and salmonella

2 but relatively little about most other phages -- and

3 also, really to be able to work in these ESKAPE

4 pathogens and more tools to be able to do molecular

5 biology in wild clinical strains, rather than just

6 model strains. There needs to be some more guidance, I

7 think -- and this is the purpose of this workshop, to

8 address issues around what are the production

9 standards, contamination profiles, how you define

10 product identity in this case, and how you develop IP

11 protection and that IP protection may actually help get

12 more investment in the field and, finally, just to

13 address that policy and economic landscape, really to

14 make actually any infective development just attractive

15 in general or to have it -- make it happen some way.

16 All right. So I would like to thank the

17 organizers and thank the audience. And I'll wrap up.

18

19 PANEL SESSION WITH Q&A

20

21 DR. TARAPOREWALA: All right. So we will -- I

www.transcriptionetc.com

75

1 now invite the speakers and the panelists to come join

2 us here in the front. We'll take a few questions from

3 the audience. And we may take a few questions online

4 if there are any. And please feel free to state your

5 name and affiliation. And also, keep your questions to

6 the point so that we stay focused on the topic of the

7 session, which is manufacturing.

8 Okay. So we have the speakers, and then we

9 have Dr. Roger Plaut from the Office of Vaccine

10 Research and Review and Ms. Anita Richardson from the

11 Office of Compliance and Biologic Quality, all in CBER.

12 And I don't see any questions and none online as yet.

13 So I will take the first stab.

14 So I was wondering if -- we talked about the

15 challenges. And I was wondering if maybe we could

16 discuss what are the advantages of -- what have we

17 learned from the rare and orphan disease drug product

18 development that can be directly applicable to

19 individualized therapeutics?

20 DR. GAO: So I believe the experience from the

21 ultra-rare disease or rare disease gene therapy can be

www.transcriptionetc.com

76

1 very helpful or informative to developing the

2 individualized therapy because I think, if you think

3 about the process of discovery or develop gene therapy

4 product -- it doesn't matter for ultra-rare, or rare,

5 or moderate-rare disease, or prevalent diseases -- I

6 think should be the same. And actually, the good thing

7 is, unlike large applications such as Duchenne’s

8 muscular dystrophy, each patient may need several, 10

9 to 15 or even up to 10 to 16 dose, vector dose, per

10 patient. But for those rare diseases, the population

11 is much bigger -- much smaller, and particularly ultra-

12 rare.

13 So the manufacturing burden is less. I think

14 current technology should be able to apply for

15 manufacturing. So that's advantage.

16 DR. TARAPOREWALA: Anybody else who would like

17 to take a stab?

18 DR. GILL: I have a question for Dr. Gao

19 actually. So for these ultra-rare diseases, is the

20 manufacturing mostly just lab scale at that point, or

21 are you still finding a CRO to try to make this stuff?

www.transcriptionetc.com

77

1 DR. GAO: Yes, this is a very interesting

2 question. Based on our experience as a gene therapy

3 center at UMASS and Rare Diseases Institute, so far, I

4 think for those kind of few patients, single patients,

5 or a couple patients, even academic viral vector core

6 should be able to address the needs. And we have been

7 very much enjoying this kind of collaboration with

8 academic GMP manufacturing core. I will say it's quite

9 -- much easier than bigger trials and bigger disease

10 population.

11 DR. TARAPOREWALA: We'll take one from the

12 audience.

13 GUY: So my name is Guy (phonetic). And I

14 don’t know if you can hear me. Is the microphone

15 working?

16 UNIDENTIFIED MALE: This one's working. You

17 can come back here if you want.

18 GUY: Oh, okay.

19 UNIDENTIFIED MALE: Just come back here. Use

20 this one.

21 GUY: Okay. It’s an individualized microphone

www.transcriptionetc.com

78

1 here. All right. So this is a question direct to

2 Professor Gao. You mentioned that manufacturing

3 capacity is clearly the challenge to produce AAV.

4 Obviously -- and if we can actually create a cell line

5 that can produce high titers of the AAV, you don't have

6 to do the transfection. So what exactly the challenges

7 are they and to produce such a cell line, if we

8 actually can do that and the manufacturing capacity

9 should be easily overcome? So I'd like to hear your

10 opinion on that.

11 DR. GAO: Thank you. I think you refer to AAV

12 production. So the major issue is that it's we need

13 regulatory protein and a capsid protein for

14 manufacturing process, particularly REP, regulatory

15 protein. That protein is very cytotoxic. And the

16 capability to generate stable integrated and producer

17 cell line, introduce the REP express under controllable

18 levels, particularly inducible levels, it has been a

19 major challenge. So that could be one issue.

20 The second issue is that other than

21 replication biology and packaging biology, AAV very

www.transcriptionetc.com

79

1 much depends on cellular factors to -- actually,

2 cellular factors contribute to all this replication and

3 packaging process. Our understanding of the cellular

4 factors that can enhance or improve gene therapy vector

5 production, we do not understand. I think if we can

6 understand the REP, where you establish your stable REP

7 cell line, as well as understand the cellular factors

8 and, importantly, if we can come up much tighter

9 inducible system that you can pharmacologically or some

10 other ways induce regulatory protein expression, that

11 will significantly give us opportunities to generate

12 so-called real producer cell line.

13 DR. TARAPOREWALA: Thanks. We can take the

14 next question from the audience.

15 MR. ALDRICH: Hello. Good morning. My name

16 is Steve or Stephen Aldrich. I'm 64 years old. I'm

17 the founder of a company called MyCancerDB. I am a

18 stage-4 adenocarcinoma of the esophagus patient.

19 I was diagnosed in March, late March of 2017.

20 For those of you who aren't familiar with that, most

21 people die for sure within two years. It's a very,

www.transcriptionetc.com

80

1 very deadly cancer.

2 Here I am three years later. Part of the

3 reason is that I was able, being a Harvard-trained

4 biologist, to recognize that if there was an answer to

5 my problem , because the prognosis is -- or the

6 prescriptive care is strictly palliative, that it was

7 going to be found in my data. And so I had -- was very

8 fortunate to be able to have all of my fundamental

9 sequencing data, my cancer genome, my healthy genome,

10 my microbiome, et cetera, and the related datasets,

11 done. And with that information, I was able to

12 leverage it to, first, identify a clinical trial that

13 kept me alive for a while.

14 And while that was going on, I had designed,

15 tested, and manufactured a fully personalized

16 neoantigen peptide cancer vaccine. And as a

17 consequence of going through that, I realized that the

18 current system was broken and that we needed to be able

19 to accelerate access to this kind of fully personalized

20 N of 1 therapy to the 1,700,000 patients who were

21 diagnosed with cancer in the U.S. last year and

www.transcriptionetc.com

81

1 especially the 610,000 that died.

2 So a couple of points I want to make about the

3 panel. The first is that, in order to enable the kind

4 of gene therapy that was presented, we have to

5 completely reengineer the entire system. And that

6 reengineering starts with the fundamental genetic data

7 of the patient, the sequencing data.

8 Clearly, to all your presentations, it's the

9 sequencing data that's fundamental. Whether it's for

10 phages or for people, it's the sequencing data that we

11 must have. But who controls that data?

12 If that data is the most important

13 personalized information about an individual, shouldn't

14 we create an infrastructure that keeps control and

15 ownership of that data with the individual? I just

16 thought I'd bring that to the panel and ask them for

17 comment. I think what we're doing at MyCancerDB is

18 putting in place that infrastructure.

19 And the idea is to be able to leverage that

20 information to virtualize the supply chain in a way

21 that shrinks the cost, speeds up the development

www.transcriptionetc.com

82

1 process, and maybe does something about the 610,000

2 people who died of cancer. So I -- last year, which is

3 12 times the number that died in the Vietnam War. I'd

4 be interested in your comment.

5 DR. TARAPOREWALA: So I guess that was a

6 comment, maybe not a question. And if nobody in the

7 panel has a comment towards that, we might move on to

8 the next question, if there's one from the audience.

9 MR. CAMPBELL: Hi. This is Joe Campbell from

10 NIAD. I have a question for Dr. Gao. I was wondering

11 -- you mentioned the advantage of flipping the ratio

12 from 90 to 10 to 10 to 90. And that obviously would be

13 a great advantage. And I'm just wondering if you have

14 any thoughts on whether -- what's likely to be

15 important, the vector, the payload, or the strain in

16 which you grow it in? And I guess one further

17 question. Have you ever, thought about making the

18 unfilled vector have something like an endonuclease, a

19 very rare endonuclease cut site that the filled one

20 wouldn't have, so that you could select against it?

21 DR. GAO: Very interesting questions. So this

www.transcriptionetc.com

83

1 flip from 90 to 10 to 10 to 90, it's quite challenging.

2 Actually, if you ask me the answer, I don't have the

3 answer. But I know where to go.

4 That is what I just talked to the first person

5 who asked the question from [inaudible]. It's the same

6 issue. It’s we have to understand how AAV replicates

7 and packages.

8 And we have to understand how cellular factors

9 can help us. Because AAV itself, it's almost a virus.

10 It needs a lot of things from host and from the helper

11 virus. So that is definitely one thing to consider.

12 Second thing is the purification methods. If

13 we are defective -- I mean virus is defective to

14 packaging for particles. But if we have a scalable

15 GMP-compatible process to separate empty from full,

16 then we should be able to enhance the potency of our

17 drug, reduce immunogenicity of those useless empty

18 particles.

19 DR. TARAPOREWALA: Okay. Thank you. We'll

20 take the next question from the audience.

21 MR. KELLY: Good morning. My name is Matt

www.transcriptionetc.com

84

1 Kelly from Sarepta Therapeutics. I just had a follow-

2 up question on the empty/full ratio.

3 DR. TARAPOREWALA: Could you speak up a little

4 bit because we cannot hear you.

5 MR. KELLY: I just have a follow-up question

6 on the empty/full ratio. As Dr. Gao highlighted, a 90

7 percent full count would be acceptable. From an FDA

8 perspective, a commercial scale, is this also an

9 acceptable reality for an approval?

10 DR. TARAPOREWALA: So I think at this point,

11 we are taking questions to see what the researchers

12 think in how to develop individualized therapeutics.

13 And I think Dr. Gao just mentioned that the empty

14 ratios does affect the potency and does affect the

15 purity and the immunogenic load. So I think that

16 applies rather product is in clinical trial or being

17 developed for licensure. So I think his answer is

18 pretty broad and comprehensive in that respect.

19 MR. KELLY: Okay. Thank you.

20 DR. TARAPOREWALA: We'll take the next

21 question from the audience.

www.transcriptionetc.com

85

1 MS. HESTERLEE: Hi. This is Sharon Hesterlee

2 from the Muscular Dystrophy Association. So I

3 appreciate very much all the improvements in process

4 development that have helped increase the yield in

5 manufacturing. But I'm wondering if we're going to hit

6 a limit at some point where we really can't further

7 increase the yield in a practical way. And should we

8 be looking at things more like targeting? You

9 mentioned early on that that was one of the issues.

10 For example, AAV9, if you're trying to target

11 the brain, the vast majority, if you go systemic, is

12 still going to liver. So I could see that you might

13 get an order of magnitude improvement if you could just

14 better-target vectors rather than increase

15 manufacturing. Is that something to consider?

16 DR. GAO: Yeah. That is a great question.

17 Actually, that's the whole field of gene therapies and

18 particularly AAV gene therapies. We have been

19 struggling for 50 years. That's what we're trying to

20 do.

21 But what I have to tell you is I think recent

www.transcriptionetc.com

86

1 research demonstrated what is achievable. I refer to

2 Dr. Ban Deverman's publication in Nature Biotech a

3 couple years ago. I think he demonstrated that you can

4 accomplish that more efficient cell type, tissue type,

5 targeting. But the only issue we're running into now,

6 soon afterwards, we realized what you select against

7 will be what you get out of the selection system.

8 So previously, the selection was done in

9 mouse. And we generated the super mouse CNS vectors.

10 So the next stage, I think as was discussed by Dr.

11 Marks, that we look into potential humaned mice, as

12 well as probably primates, animal models, to do that

13 selection. I think that people know how to do it, just

14 have to switch the system.

15 MS. HESTERLEE: Thank you.

16 DR. TARAPOREWALA: We'll take the next

17 question from the audience again.

18 MR. MCFARLANE: Thanks. Richard McFarland for

19 the Advanced Regenerative Manufacturing Institute. So

20 as Dr. Gao said for the hyper-rare diseases, you think

21 maybe laboratory scale manufacturing would be

www.transcriptionetc.com

87

1 sufficient. It seems that raises -- we've done a lot

2 of talking about traditional CMC, but it seems that

3 also raises questions about distributed inspection and

4 regulatory compliance for those centers. And I wonder

5 if you can share any thinking about what you've had

6 about how you're going to inspect those facilities if

7 they do exist across multiple hospitals.

8 DR. TARAPOREWALA: You can go ahead, and then

9 Anita can weigh upon it. Go ahead, Dr. Plaut.

10 MS. RICHARDSON: Richard, thank you for that

11 question. And I'm not sure I have a simple answer. I

12 think that we are looking at those types of facilities

13 on a case by case basis, taking all the facts and

14 circumstances and the product into consideration and,

15 also, the flexibility needed in this field.

16 MR. MCFARLANE: Well, Anita, I'm glad you

17 don't have a simple answer because I don't think it's a

18 simple solution. But I think it's something that I

19 know we're thinking about and I think others may be

20 thinking about. So maybe it's a place for focus

21 workshops or guidance, something in the future to help

www.transcriptionetc.com

88

1 think through those issues as you go from an IND to a

2 true manufacturing facility.

3 DR. TARAPOREWALA: Okay. We'll take the next

4 question from the audience again.

5 MS. GALEMBO: Marian Galembo from BiomX

6 (phonetic). And I will talk about phage therapy. So

7 Dr. Gill has really put forward a whole lot of the

8 dilemmas and the problems that we may be facing with

9 individualized therapy and manufacturing in hosts that

10 could be derived from individual patients and each one

11 with different characteristics. I would like to hear a

12 bit about the Agency's approach on how to handle those

13 differences and variations in terms of impurity

14 characterization, if there is any thoughts about that.

15 DR. PLAUT: So the goals of this workshop are

16 to get feedback from the community, from stakeholders.

17 So we are not really prepared to make any public

18 statements about our -- how we're regulating these

19 products or how we intend to regulate these products.

20 I can tell you that every decision we make is based on

21 science, based on evidence. And we ask sponsors or

www.transcriptionetc.com

89

1 potential sponsors to come to us and provide all their

2 data and to propose to us what they think their

3 specifications should be, what they think the limits

4 should be.

5 And then we have a discussion with the

6 sponsor. This is one reason why we think it's very

7 important that sponsors interact with us as early in

8 the process as they can. We're happy to have these

9 sorts of discussions in -- early on in development.

10 But again, I think it's important for you to understand

11 that we have an open mind, and we make our decisions

12 based on science and evidence.

13 MS. GALEMBO: Thank you.

14 DR. TARAPOREWALA: Thank you. We'll take the

15 next question from the audience again.

16 MS. WALKER: Hi. My name is Karen Walker.

17 I'm from Genentech. I want to thank you both for such

18 good talks.

19 I actually have a question for both of you.

20 In each of your cases, you talked about manufacturing

21 challenges from an intellectual property perspective

www.transcriptionetc.com

90

1 and a capacity perspective. And Dr. Gao, you talked

2 about the large role that CGMOs are playing in the

3 field.

4 And given that these are individualized

5 therapies and given that manufacturing platforms and

6 establishment of these platforms would drive down cost,

7 what are your ideas about how we could open up access

8 to those platforms so that we could really drive

9 targeted individualized therapy treatments to patients

10 with predictable outcomes?

11 DR. GAO: So I have to say, not myself, but

12 two weeks ago, NCATS and P.J. Brooks, and other

13 colleagues from NIH, they held a meeting on AAV gene

14 therapy manufacturing. And that meeting, their whole

15 purpose was really understand the landscape, understand

16 the challenging, understand to kind of brainstorm this,

17 all the scientists, and regulator and administrator to

18 figure a way to ask -- to address that question. So I

19 think I don't know whether P.J. is still here or not.

20 He was here in the morning.

21 They do have actually, in after reading

www.transcriptionetc.com

91

1 communications, I think they gradually -- is in the

2 process of formulating some kind of solutions for

3 ultra-rare, rare, giving these individualized

4 therapies.

5 DR. TARAPOREWALA: Dr. Brooks, would you like

6 to comment on that?

7 DR. BROOKS: Yes. Thank you for that. Thank

8 you, Guangping. At NCATS, we are certainly interested

9 in these kind of broad issues and making these

10 platforms more widely available for these types of

11 treatments. And, specifically, our -- the Cures

12 Acceleration Network, which is part of the NCATS

13 Council, is very much focused on addressing this issue.

14 And part of the goal of the meeting that we

15 had a couple weeks ago was to identify the

16 opportunities that might be available to make certain

17 investments in research approaches to really bring this

18 forward. So it is something that is under active

19 investigation, by NCATS as well as with other -- our

20 other partners within the NIH and industry, and of

21 course the FDA as well.

www.transcriptionetc.com

92

1 MS. WALKER: Thank you. And just maybe a

2 comment. I think this also speaks to the fourth bullet

3 point on the slide, which is it would be interesting

4 for FDA to think about additional guidance on how we

5 could have regulatory flexibility around establishing

6 production platforms for these individualized

7 therapies, which then would change the focus of the

8 review more to the therapy itself and the safety and

9 efficacy of those, where we could maybe control some of

10 the variability around the platforms.

11 DR. BROOKS: That issue might come up later on

12 in the afternoon as well.

13 MR. ALDRICH: Can I just follow up that

14 excellent question and the point? There was actually a

15 question in my earlier comment. And it has to do with

16 -- for the reengineering that we're going to have to do

17 around manufacturing, there are methodologies that are

18 well-known to the FDA that are used in the regulation

19 of, for example, the food industry, where you use

20 hazard and critical control-point methodologies to

21 regulate, in a scientific way, the safety of a

www.transcriptionetc.com

93

1 particular supply chain.

2 And I -- that is something like that, where we

3 substitute the regulation of the end product as a

4 standard product, with the regulation of the process

5 that we use to get to that end product, is inevitably

6 the endpoint, in my opinion, of enabling N of 1

7 therapies to come to fruition. And this isn't a

8 hypothetical issue for me. This is a very concrete,

9 real issue for cancer patients everywhere, when it

10 comes to something like access to fully personalized

11 neoantigen peptide cancer vaccines.

12 So I'd just like you to comment on -- perhaps

13 our two representatives from CBER -- could comment on,

14 is there an awareness within the FDA of this idea of

15 using well-known GMP and HASIP-related methodologies to

16 -- as a basis for approving an “N of one” manufacturing

17 process, and if not, why not?

18 DR. PLAUT: So again, I will say that we are

19 openminded about these different approaches to these

20 kinds of therapies. And we see that the IND process,

21 as it stands now, is appropriate for the process for

www.transcriptionetc.com

94

1 getting products into the clinic. For as far as how a

2 product or a process could be -- could lead to

3 licensure, that is something that will remain to be

4 seen. We don't really have any sort of policy

5 statement to make at this point. But again, we have an

6 open mind about these issues.

7 DR. GILL: I'd just like to make a point. So

8 on the phage side, that's sort of -- I think what we're

9 looking at, is that you have to have kind of the

10 process approved rather than individual phages.

11 Because at least the way the field is moving right now,

12 if you're looking at, say, a personalized therapeutic,

13 say, for Acinetobacter baumannii, there's going to be a

14 collection of 200 phages or 500 phages. You're not

15 going to have detailed preclinical and clinical data on

16 all of those phages. You're going to have maybe on the

17 subset of those.

18 And then the rest of them are going to be kind

19 of carried along, based on some criteria that are set -

20 - that they look like the phages that work and so on.

21 Because I think if you require that every phage goes

www.transcriptionetc.com

95

1 through a full clinical evaluation, then it's really

2 never -- I don't think it's ever going to happen

3 because you can't run clinical trials on 200 individual

4 products.

5 DR. TARAPOREWALA: In the interest of time,

6 because we have a very good number of sessions, I urge

7 you to -- the two attendees that have gotten up to ask

8 questions, I'd urge you to get hold of the speakers

9 offline -- they will be around here for the whole day -

10 - and perhaps ask the question. So in the interest of

11 time, let's thank the speakers and the panelists for a

12 very nice session. Thank you.

13 DR. RAYCHAUDHURI: So I'd like to thank all

14 the speakers, the panelists, and the audience, for a

15 stimulating discussion. And I'm really sorry we had to

16 cut it short. But please do take advantage of the

17 speakers being here to speak with them one on one.

18 We'd like to now take a short, maybe 10-minute

19 break, just to stretch your legs, and we will reconvene

20 with session 2. Thank you.

www.transcriptionetc.com

96

1 [BREAK]

2

3 SESSION 2: TOOLS FOR SAFETY TESTING AND DEVELOPMENT

4

5 DR. RAYCHAUDHURI: Okay. Could everybody

6 please take your seats because we'd like to get Session

7 2 started? So that was a short break. And I hope you

8 had a chance to stretch your legs. We are ready to

9 start Session 2.

10 I'd like to introduce Dr. Sandhya Sanduja, who

11 will moderate the session. Dr. Sanduja is acting team

12 lead for Pharmacology Toxicology Branch I in the

13 Division of Clinical Evaluation, Pharmacology, and

14 Toxicology in Office of Tissues and Advanced Therapies.

15 Dr. Sanduja will moderate this session, which is on

16 nonclinical evaluation and tools for safety testing for

17 individualized therapeutics.

18

19 SESSION 2 MODERATOR INTRODUCTION: DR. SANDHYA SANDUJA

20 (CBER)

21

www.transcriptionetc.com

97

1 DR. SANDUJA: Thank you, Gopa, and thank you,

2 everybody who's present here as part of our workshop to

3 facilitate end-to-end development of these

4 individualized therapies. So after a very interesting

5 and engaging Session 1 on manufacturing of these

6 individualized therapies, it is my pleasure to take you

7 to our Session 2, which will focus on tools for

8 preclinical testing of these individualized therapy

9 products that are regulated by CBER. The session will

10 begin with presentations by our distinguished speakers,

11 Dr. Albert Seymour from Homology Medicines, Dr. Malachi

12 Griffith from Washington University School of Medicine,

13 and Dr. Keith Joung from Harvard Medical School. These

14 presentations will be followed by a panel discussion,

15 which will be led by our speakers and our CBER subject

16 matter expert, Dr. Zuben Sauna, and our audience.

17 All right. So to give you a high-level

18 overview for this session, let me begin with this

19 schematic, which shows the traditional drug development

20 pathway. The preclinical testing paradigm for

21 individual therapeutic products looks very similar to

www.transcriptionetc.com

98

1 these other therapeutic modalities, where efforts are

2 spent during the discovery phase. They focus on

3 identification and optimization of the lead candidate,

4 followed by IND-enabling preclinical studies that

5 evaluate and establish proof of concept and safety of

6 these products.

7 However, when we talk about individualized

8 therapeutic products, the key distinction -- obvious

9 distinction that these products have are that they are

10 patient-specific. They are patient-tailored products,

11 and testing a healthy individual is either not feasible

12 or not ethical. So this is where the standard paradigm

13 of drug development won't apply.

14 During our session, we will hear from each of

15 our speakers some of the key challenges that are

16 encountered during preclinical safety testing and

17 development of these individualized therapeutic

18 products. We'll also hear from them what are some of

19 the opportunities we have to address these challenges,

20 including some of the regulatory -- the existing

21 regulatory framework to address these challenges, and

www.transcriptionetc.com

99

1 these challenges and opportunities, how they are

2 perceived by the developers as well as regulators. So

3 as we know, these products are patient-specific. These

4 are tailored for either one or a small number of

5 patients, instead of being tested in healthy

6 individuals.

7 So this brings in an inherent challenge

8 associated with these therapeutic products. And what

9 we expect from a preclinical testing program for these

10 individualized therapy is, that since these are going

11 into one or a small set of patients, to strike a

12 balance or to strike that favorable benefit-risk

13 profile, preclinical evidence to support the rationale

14 for safety of administration of these products and

15 patients. Such support must come from one or more

16 preclinical studies that are conducted in a relevant

17 animal model of disease using the intended route of

18 administration, since for these individualized

19 products, many times, these can be rare or ultra-rare

20 diseases, where the only patient population is

21 pediatric and in those cases, preclinical studies need

www.transcriptionetc.com

100

1 to establish prospect of direct benefit, as is required

2 by Code of Federal Regulation.

3 So fulfillment of all these parameters can get

4 extremely challenging in the absence of a relevant

5 animal model of disease. When there is actually a

6 relevant animal model of disease, there may be other

7 challenges associated with preclinical development of

8 these products.

9 And these challenges may vary. They may arise

10 on a case by case basis and may include appropriate

11 dose-level extrapolation, delivery with respect to the

12 intended clinical route of administration, the

13 procedure and device, as well as informing the

14 appropriate clinical monitoring for the clinical trial.

15 For in silico or computational tools that are used for

16 preclinical testing, there are challenges that lie in

17 validation of these algorithms to test -- and other

18 test methods for robustness, whether they can

19 confidently and reproducibly perform to the desired

20 standards of safety and activity of these personalized

21 therapeutic products.

www.transcriptionetc.com

101

1 Some additional challenges, which can arise

2 later in development, may include additional -- conduct

3 a requirement of additional nonclinical studies that

4 may be needed, depending on if there are significant

5 manufacturing changes that happen during later part of

6 development, also for the potential for development and

7 reproductive toxicities that may be present. So these

8 challenges, which may actually seem as hurdles to begin

9 with, may not necessarily be roadblocks to development

10 of these individualized therapeutic products. Instead,

11 they may be taken as opportunities which can drive

12 innovation, so particularly, with these products, where

13 we know there is so much science-based evidence in

14 development.

15 So they can drive innovation, particularly

16 advancement, in science and technology for better

17 models and approaches to preclinical testing. For

18 example, in silico methods that are used to inform

19 safety and activity of products can have adaptive

20 design to them. And as our experience -- clinical

21 experience grows with these products, that adaptive

www.transcriptionetc.com

102

1 design can be -- is incorporated in the tools that are

2 used to inform safety and activity.

3 Similarly, innovation happens when novel in

4 vitro methods are being developed. There can be -- we

5 have seen examples where patient-derived IPSCs or

6 patient-derived organoids can be used, particularly

7 when a relevant animal disease model is not available.

8 And there, such in vitro models can actually allow for

9 reduction, refinement, and replacement of animal

10 testing. Innovation can also play a role in developing

11 relevant and more robust in vivo models for testing

12 safety and activity of these products.

13 Next is collaboration. These hurdles can

14 actually be instrumental in driving collaborative

15 efforts, knowledge sharing in the community. So

16 instead of reinventing the wheel, as Dr. Marks alluded

17 to earlier, every single time, especially when using a

18 similar vector or similar platform technologies,

19 stakeholders may have the opportunity to leverage

20 existing preclinical and clinical data. That way, we

21 are harmonizing our efforts and leveraging data that's

www.transcriptionetc.com

103

1 already available to facilitate a faster translation to

2 the bedside.

3 And finally, these hurdles are instrumental

4 again in driving engagement with regulators, in

5 basically interactions with regulators to discuss the

6 preclinical program and how we can all agree and come

7 to terms for a feasible path forward for a specific

8 product. So with our session -- as we start the

9 session, after hearing from our speakers, I think we

10 all will be engaging in a discussion where the key

11 points to discuss would be what are some of the

12 challenges that are associated with preclinical testing

13 and development of these products including: platform-

14 based versus any product-specific concerns, use of

15 computational tools in safety testing of these

16 products, how leveraging existing nonclinical data

17 across similar products can be done, and regulatory

18 approaches and flexibility to preclinical testing of

19 these products.

20 So without any further delay, I would like to

21 welcome our first speaker, Dr. Albert Seymour. He's a

www.transcriptionetc.com

104

1 chief scientific officer of Homology Medicine, a

2 company that's developing gene therapy and gene editing

3 technology to treat patients for rare diseases. He has

4 a Biology from University of Delaware, an MS in

5 Molecular Biology from Johns Hopkins University.

6 And he received his PhD in post-doc training

7 in human genetics at the University of Pittsburg. He

8 has more than 20 years of experience in taking human

9 genetics to pharmaceutical RND, resulting in delivery

10 of multiple therapeutic programs into development. So

11 we welcome you and looking forward to your

12 presentation.

13

14 PRECLINICAL APPROACHES/CHALLENGES IN DEVELOPMENT OF

15 INDIVIDUALIZED THERAPEUTICS - DR. ALBERT B. SEYMOUR

16

17 DR. SEYMOUR: Thank you. Thanks, everybody,

18 and first, thank the FDA for giving me this opportunity

19 to come and talk a little bit about some of the

20 preclinical approaches and challenges to developing

21 these individualized therapeutics. I have to admit,

www.transcriptionetc.com

105

1 when I got the invitation, I really sat and gave it a

2 lot of thought, working with my colleagues as well, for

3 what are some of these challenges? Because we've been

4 focusing a lot on bringing therapies to rare, ultra-

5 rare disorders. And so I think a lot of those unique

6 opportunities and challenges fall also within very

7 individualized therapies.

8 And so I sort of pulled a collection of these

9 together. So one of the things I looked at right away

10 is some of the work that actually came out of Dr. Yu’s

11 lab out of Children. And this was -- really got a lot

12 of press. But it really exemplified that, where

13 there's a need, there is an opportunity to bring these

14 forward. And this was when publishing in New England

15 Journal of Medicine with this patient-customized

16 oligonucleotide therapy for a young child with a form

17 of Batten's disease.

18 And in ten months, going from sequencing,

19 identifying the unique mutation within that patient to

20 having an oligo that could actually correct that to

21 them being able to treat that patient, was just

www.transcriptionetc.com

106

1 fascinating to actually read and see how they were able

2 to go through that in leveraging some of the aspects

3 around cell-based models from that individual patient

4 because there wasn't an in vivo model. But then you

5 could also pull some information that was also

6 available on just the chemistries of the unique

7 oligonucleotides.

8 So with that, there are a lot of platform data

9 that can be leveraged to improve the efficiency. But

10 as we were thinking about this, there are really three

11 buckets that go into this, that the fundamentals of

12 benefit and risk assessment must still apply. We are

13 still bringing therapies forward to treat human

14 patients.

15 The other aspect is -- you heard from some of

16 the earlier speakers, so Dr. Gao, and then some of the

17 work that we're doing at Homology, there’s single

18 administration versus chronic dosing. So something

19 like AAV, it's a single administration. It brings, I

20 think unique challenges to an individualized patient

21 because of the ability you really can only dose once.

www.transcriptionetc.com

107

1 You really have to get it right that first time versus

2 something that you can do with chronic dosing. Perhaps

3 you can come in with a much lower dose and work your

4 way up to look for activity.

5 And then finally, with that, there are very

6 modality-specific requirements. And I'll walk through

7 some of these and exemplify these as I go along. So as

8 I think about these considerations, establishing the

9 fundamentals of benefit and risk, I really broke it

10 down into two categories, product-specific -- and what

11 I mean by that is that the actual -- if you're going to

12 replace the gene, the actual molecule that you're going

13 to replace versus a platform. So this would be, say,

14 if you're looking at a capsid AAV9, AAV15, AAV2 -- that

15 could be, can you leverage some of that that's more

16 platform-driven.

17 So from a product-specific perspective, you

18 still have to establish some form of biological

19 plausibility. This could be proof of concept in a

20 model, if one exists. But a lot of times, the timeline

21 that you need is not there to develop a new model. So

www.transcriptionetc.com

108

1 you may have to go to more in vitro models.

2 You also have to think about projecting the

3 dose for the clinical testing. Can you establish a

4 minimal efficacious dose? I think a really important

5 aspect is to identify dose response relationship early

6 and establish that pharmacology, particularly as you're

7 moving into that patient because a lot of these

8 patients may not have characterized natural history.

9 So you're going to want to know, if your drug gets into

10 that patient, are you actually engaging with the target

11 and seeing a pharmacology that you would be expecting.

12 And then finally, the safety margin would be very

13 product specific.

14 From a platform perspective, establishing the

15 delivery vehicle biodistribution. So there are a lot

16 of studies out there that perhaps you could cross-

17 reference if existing biodistribution are using AAV

18 capsid, for example. If you have biodistribution of

19 the capsid itself, say, in non-human primates or other

20 relevant species, can you leverage that when you come

21 in with another product but using the same AAV-based

www.transcriptionetc.com

109

1 capsid delivery?

2 And then there are also other aspects around

3 class-specific effects. So with the oligonucleotides,

4 the example that I just brought up from Dr. Yu at the

5 Boston Children's with the Batten's patient, really

6 looking at the underlying backbone of the chemistry.

7 Can you leverage what was already out there to then

8 bring that forward to help eliminate or reduce some of

9 the risk that may be associated with moving that

10 forward?

11 So upon establishing biological plausibility,

12 it's really characterizing the impact on this. And

13 this, I think, is really unique to the specific product

14 on that biological plausibility. What I mean by that,

15 if we're going after a monogenic disorder, we know what

16 the mutation is. We may be able to characterize that.

17 Are there cell lines that harbor that patient-specific

18 mutation? And ideally, depending on the therapy, is

19 there an animal model of that particular disease?

20 Because there's several things you'd want to

21 test: will the therapy reverse the effect of the

www.transcriptionetc.com

110

1 mutation? Will we see an increase in a protein

2 activity for something where that activity is missing?

3 Or can you decrease that activity in something that has

4 what's called a gain of function type of mutation?

5 So utilizing that to characterize the activity

6 establishes the dose response, understand durability of

7 that. And then I think one thing I really want to

8 bring up is establishing these endpoints that perhaps

9 you can test preclinically, that are the same aspects

10 and the same endpoints that you would test clinically

11 as you move into that, even into an individualized

12 patient. I want to use here just one -- an example of

13 that and establish a biological plausibility.

14 Here's the gene therapy for a disease called

15 PKU. There fortunately is a mouse model for this

16 disease. The mouse is missing or has a mutation in the

17 same enzyme that causes a human disease, phenylalanine

18 hydroxylase.

19 In this mouse, when you have two missing

20 copies of that or two mutations on that, you -- that

21 mouse cannot metabolize phenylalanine, very similar to

www.transcriptionetc.com

111

1 the human condition. So we can use that mouse to

2 actually do a single administration, and then see not

3 only do we reduce phenylalanine, but we can see that

4 loss durably over the lifetime of the mouse. So this

5 really helps establish the biological plausibility of

6 that specific product. We're adding back in

7 phenylalanine hydroxylase. We see it in vivo in an

8 animal model.

9 Here's another example, trying to understand

10 the dose relationship. So this is in a different

11 disease. This is a disease called metachromatic

12 leukodystrophy. It's a rare genetic disorder of

13 lysosomal storage.

14 These patients are missing an enzyme called

15 arylsulfatase. And unfortunately, when they're missing

16 that enzyme, that enzyme typically works in a lysosome

17 that is responsible for breaking down a product called

18 sulfatides. What happens in these children is these

19 sulfatides increase, and that increase in sulfatide is

20 toxic to the cells.

21 We can reference human data from people that

www.transcriptionetc.com

112

1 are out there that have roughly 10 to 15 percent of

2 normal human ARSA activity. There's a common

3 polymorphism within the ARSA that results in that.

4 These people generally are normal. And so we know that

5 at least if we can hit a 10 to 15 percent of a normal

6 activity, that should get us into something that would

7 be therapeutically relevant. And what you see on the

8 right is looking in a knockout model of that, showing

9 that as we increase doses with those, we can start to

10 see an increase in that activity in the brain trying to

11 shoot for that target threshold of 10 to 15 percent,

12 based on human data.

13 The other aspect is really trying to project a

14 starting dose for the clinical administration. So

15 again, this is unique to the specific product, but

16 there might be some platform-specific challenges and

17 opportunities to overcome that. So using AAV

18 specifically, AAV really does require in vivo data to

19 project a dose. You see differences in how AAV works

20 from an in vitro to an in vivo setting.

21 So here, you can either use an animal model of

www.transcriptionetc.com

113

1 human disease, if one exists. That will allow you to

2 assess the effect on the actual underlying mutation

3 that's similar to the example that I just gave on

4 introducing phenylalanine hydroxylase into ENU 2 mouse

5 model of PKU. So you can assess either an increase in

6 activity there or a decrease in vivo.

7 However, if you don't have an animal model, at

8 least of the things you can do is look in referencing

9 different data. So if you're using a specific delivery

10 approach such as AAV, if we already have that data from

11 other studies, can we reference that? Because that

12 will give us a sense of different doses, how many

13 vector genomes, at least we get from the capsid,

14 delivery into the liver, into crossing the blood/brain

15 barrier into the CNS. Can we leverage that to move

16 those forward?

17 So along these lines, again, unique to the

18 specific product, platforms that require repeat dosing,

19 you can utilize these models either in vitro or in vivo

20 to establish a dose response, to define that minimal

21 efficacious dose, and perhaps to develop biomarkers

www.transcriptionetc.com

114

1 indicative of pharmacodynamic activity, meaning that

2 your drug is engaging with the target and then having

3 the effect pharmacologically that you were expecting

4 coming in. For single administration platforms,

5 meaning a single dose, you can do -- all the first

6 three are the same. The only thing is for that fourth

7 that's very unique for these is that establishing the

8 starting dose that has a high likelihood of benefit.

9 Because particularly, with AAV or other types

10 of these approaches, the ability to re-dose at this

11 point is challenging. And so if you're coming into an

12 individualized person, you really want to make sure

13 that that first dose is going to have the activity that

14 you want. So that sort of raises the bar a little bit

15 form an AAV perspective. But the opportunities are

16 there, I think, to utilize different models to begin to

17 address that.

18 Just to exemplify that, again, I show a lot of

19 PKU data because we have a program that just is in

20 early clinical programs right now for that. Here's

21 some of the data from a 28-day dose ranging finding

www.transcriptionetc.com

115

1 study in ENU 2 mice. And so what we're able to show is

2 characterizing both males and female mice, looking

3 across a variety of doses, looking at a reduction in

4 phenylalanine. So that's a direct measure of

5 increasing PAH activity. So we're putting back in

6 something that was missing.

7 We can pick a dose that identifies that. And

8 then we can also look at another marker, which is

9 called tyrosine. So tyrosine is the direct product of

10 phenylalanine metabolism via phenylalanine hydroxylase.

11 And so we have two approaches that we can look at to

12 help identify what are those doses, at least in the

13 mouse model, that may deliver the effect that we're

14 looking for.

15 And then taking that information, we could

16 actually apply some PKPD modeling. And so we can

17 utilize those models to try to get an assessment of, if

18 we go into a patient population that, say, has a

19 baseline serum phenylalanine levels of 1,200, 1,800, or

20 even all the way up to 2,400, are there doses that,

21 based on the preclinical data, would deliver that kind

www.transcriptionetc.com

116

1 of result. We can then take the modeling and identify

2 and make some predictions around what percentage or

3 proportion of subjects would we expect to have that.

4 So based on this modeling, it's based on mouse

5 efficacy data. So what's unique about these mice, they

6 are homozygous for a genotype phenotype. We know that

7 if we go into the general population of humans, they're

8 not usually homozygous for a specific genotype, so it's

9 much more heterogenous.

10 But it really can allow us to select a dose

11 range that we know would have activity. And then we

12 can align that up with what we're seeing from our GLP

13 toxicology studies to assess where is our safety

14 margin, if you will, to make sure that these can fall

15 into that. I think the translation to humans is

16 required to fully characterize these, just given the

17 inherent variability of what you would see in human

18 populations compared to what you see in a very well-

19 controlled mouse and animal model environment.

20 So then moving along is really establishing

21 that safety margin. Here's where some of the, I think,

www.transcriptionetc.com

117

1 challenges come that would be unique to the specific

2 product. I know that if you're using one particular

3 AAV, the inside of that AAV is different as you go

4 across different diseases. And we know that any kind

5 of safety effect can be both based on the delivery

6 vehicle but also based on the actual API, if you will,

7 that's going to be delivered to that.

8 So you can use a model of human disease of

9 what's the safety margin, aligned with the

10 pharmacology. That's where, if you do have a model, it

11 really gives you a lot of that information, seeing

12 assessment or pharmacology, but then you can also see

13 assessment of where that safety margin is to assess

14 that. If you don't have a model, you can still use

15 wild-type animals to get a sense of the safety margin,

16 as well as the biodistribution of the therapy.

17 And then finally here, you can pull in some

18 reference studies particularly around the delivery

19 vehicle, so whether it's an AV capsid where you can

20 utilize that or some of the known class effects. What

21 I mean by here is -- again, I will use AAV as an

www.transcriptionetc.com

118

1 example. We know that AAV, there's immune responses to

2 it, and a lot of those are cytotoxic t-cell mediated,

3 against the liver. And so one of the main tox organs

4 is the liver. So you can reference that from other

5 studies to then guide you as to what to monitor, at

6 least as you think about moving into the clinic.

7 As far as safety and biodistribution, again,

8 from a product-specific, there could be mechanism-

9 driven safety. So what are the expression levels of

10 the specific product? What are the level of inhibition

11 that you need? If you inhibit 50 percent, you might be

12 safe. But perhaps, if you inhibit too much, you might

13 have some safety consequences and vice-versa on the

14 cell-specific challenges around maybe expressing too

15 much.

16 I think Dr. Gao brought that up a little bit,

17 as one of the last hurdles in getting into the clinic

18 is trying to regulate this. Some cells, based on

19 delivery, you may hit them very hard, and then you may

20 have overexpression of your product in that particular

21 cell. And so we need to be cognizant of those aspects

www.transcriptionetc.com

119

1 as well.

2 There are also chemical or structure-driven

3 safety aspects, so off-target effects. I think we're

4 going to hear a little bit about that in two speakers

5 from now, with Dr. Joung, and then also, the

6 interaction with the immune system. I mentioned

7 briefly about AAV. We also know with other

8 oligonucleotides, other antibodies, there are just

9 unique class-specific effects that you need to take

10 into consideration.

11 As far as the platform, once you move to the

12 platform, again, you can look at the properties of the

13 delivery vehicle. So I've spoken quite a bit about the

14 specific AAV capsid and understanding how that capsid

15 delivers. The capsid is really what's delivering it to

16 the different cell types. And so whatever --

17 regardless of what you have inside that capsid, the

18 capsid itself is usually what's going to deliver it to

19 those particular cells. Or you could also think the

20 same thing with a lipid nanoparticle or an antibody as

21 you get a sense, if you're utilizing them as drug

www.transcriptionetc.com

120

1 delivery vehicles.

2 The other aspects from platforms are assays

3 for measuring. So one of the unique things that, as

4 you start to develop this, there are assays out there

5 for measuring caspids and specific antibodies against

6 the capsid. So how do you measure that?

7 If it's against a specific capsid, you should

8 be able to leverage those across many different studies

9 or even other things such as anti-antibodies. So if

10 you're coming in with a biologic and you develop those

11 antibodies, if it's the same antibody being utilized in

12 a different thing, I think you can leverage some of

13 that data going forward for those biodistribution.

14 So here's an example of biodistribution data

15 that we generated in cynomolgus monkeys, with one of

16 our capsids, AAV HSC15. So we did this in two

17 different dose. And we were able to collect a variety

18 of tissues. This data now, these data, we can then

19 cross-reference for other studies that were using AAV

20 HSC15. And it really helps accelerate some of those

21 preclinical testing that we would be looking into for

www.transcriptionetc.com

121

1 utilizing AAV HSC15.

2 So finally, I just wanted to sort of wrap up

3 with the last few slides. As we think about this,

4 different targets require both product-specific

5 nonclinical characterization, as well as platform. So

6 just the example here, if we're using an AAV HSC15, and

7 we have disease one, the AAV HSC, the capsid, is

8 exactly the same, whether we're going after disease one

9 or disease two, just to exemplify this. So we can

10 cross-reference by distribution. We can utilize capsid

11 assays. We can utilize a lot of these different

12 aspects, just thinking about moving that forward to

13 help accelerate that.

14 But the inside is very different. So we may

15 have different promotors. We may have different trans

16 genes. And that's the product-specific aspect that we

17 do have to generate some testing for in --

18 preclinically, and so really looking at establishing

19 biological plausibility. So this disease and that

20 disease will have different cell-based models, perhaps

21 different animal models that we still have to go

www.transcriptionetc.com

122

1 through and assess, dose selection, and then finally

2 the underlying safety margin.

3 And then I talked a little bit about this

4 before. But the class effects can be informative but

5 perhaps not sufficient for a total safety assessment.

6 But we can pull that information in to utilizing as we

7 think about these. So one of the things just to

8 highlight here is just, as that data is being collected

9 and we're seeing more and more of these technologies

10 generate more data, publishing these data, you can

11 start to think about, from an individualized

12 perspective, moving those forward and using that to

13 help accelerate some of these individualized type

14 approaches.

15 So finally, platform-specific considerations,

16 in terms of selecting a dose, platforms that are

17 amenable to repeat dosing, so basically small molecules

18 and oligonucleotides, antibodies, protein replacement -

19 - a lot of those you can start at a lower dose and

20 increasing those doses with a safety margin. It really

21 gives you that opportunity to move forward with that.

www.transcriptionetc.com

123

1 And I think that's similar to what that example in

2 Batten's disease they were able to utilize that.

3 Ideally, align it with a PD biomarker because

4 we heard about this already. A lot of these

5 individualized diseases just don't have an established

6 natural history. So ideally, you want to know what

7 your drug is doing inside the patient and making sure

8 it's engaging with the target and seeing that activity,

9 so anything that can be done to spend time on

10 understanding those pharmacodynamic markers.

11 And then there also is some guidance around

12 microdosing. So maybe there's a unique opportunity

13 around microdosing options to even accelerate these

14 even further, as we think about taking these

15 individualized therapies forward. Again, coming back,

16 just to summarize on this. Platforms that are not

17 amenable to repeat dosing, so AAV, a lot of the other

18 genetic medicines, it is a single administration

19 aspect.

20 So I think the bar is a little bit higher in

21 terms that for particularly in individualized therapy -

www.transcriptionetc.com

124

1 - it’s an N of one -- getting that first dose right is

2 going to be essential for that particular patient. And

3 so really, that's where understanding the likelihood of

4 clinical benefit, understanding your preclinical dose

5 modeling data, going either from an in vitro, if that's

6 what you have, or really, pulling whatever you can from

7 in vivo to help drive that first dose. And maybe

8 there's an opportunity here, based on taking into class

9 effects. If you have the safety margin for an

10 individualized therapy, is that something to consider

11 that, instead of starting at the minimal efficacious

12 dose for that one patient depending on the benefit, the

13 likely benefit could be there? Do you start at a much

14 higher dose but still within your safety margin?

15 And then one of the things I wanted to -- as I

16 was thinking about this is putting in aspect around

17 repurposing existing drugs. This could actually be the

18 fastest because you'll probably have a host of

19 information already available to you, with respect to

20 safety profile, with respect to PD activity, to then

21 move pretty rapidly into these individualized

www.transcriptionetc.com

125

1 therapies. I think this is more of a rare exception

2 though because a lot of these individualized therapies

3 are going to be based on DNA sequence mutation and very

4 personalized. There just aren't a lot of existing

5 drugs out there that can target that. But it is a

6 possibility.

7 In terms of challenges and possibilities to

8 streamline this, I think the product-specific

9 characteristics, I think, have to be determined every

10 time. I think that's sort of par for the course. But

11 are there opportunities to streamline?

12 So one of the things I was thinking about is,

13 is there a form to share precompetitive platform data?

14 We heard a little bit earlier about IP. IP is always

15 going to be a consideration. But are there

16 precompetitive platform data? So whether it's AAV,

17 biodistribution, if someone is working on someone else

18 -- another AAV and we already -- and someone else

19 already has that data, how can we get that to share

20 particularly for a very severe disease that needs

21 therapy very quickly?

www.transcriptionetc.com

126

1 Commercial assays, specific to the platform,

2 so if we have neutralizing antibodies against certain

3 AAVs, can we share those? Can we get these out there

4 quickly? And then one of the other aspects that came

5 up around common manufacturing assays to support a

6 platform. So whether it's triple transfection and

7 HEK293s or whether it's SF9, are there different

8 aspects that we can take advantage of?

9 And then finally, I just want to end on really

10 thinking about this microdosing. Are there guidance or

11 white papers on the application that we can take

12 advantage of existing guidelines or initiatives that

13 are already there that the FDA has made available to

14 us? So microdosing in support of dose escalation for

15 individualized therapies, can we -- is there a

16 possibility to expand that?

17 And then the last thing I just want to bring

18 up is -- and it was brought up a little bit earlier --

19 the possibility and the openness of the FDA for what

20 are called Interact meetings. We found these to be

21 extremely helpful. And I think for something like

www.transcriptionetc.com

127

1 individualized therapy, this could be an avenue to

2 really start engaging with the regulators very early

3 because you can get quite a bit of feedback before you

4 start all of your nonclinical studies.

5 And so I just wanted to end on that aspect of

6 the different approaches. And again, thank the FDA,

7 thank the audience for listening to me.

8 DR. SANDUJA: Thank you, Dr. Seymour, for a

9 very interesting and exciting presentation. I would

10 now like to welcome our next speaker, Dr. Malachi

11 Griffith. Dr. Griffith is currently an assistant

12 professor of medicine and the assistant director for

13 the McDonnell Genome Institute at Washington University

14 School of Medicine.

15 Dr. Griffith completed his Bachelor of Science

16 with honors in biochemistry and biology in 2002 at

17 University of Winnipeg, followed by additional formal

18 training in computer science. He worked as a molecular

19 biologist and then as a computational biologist during

20 2003 to 2004 before beginning a PhD in medical genetics

21 and bioinformatics at the University of British

www.transcriptionetc.com

128

1 Columbia. After his PhD, he joined Washington

2 University School of Medicine in 2011.

3 Dr. Griffith's research is focused on the

4 development of personalized medicine strategies for

5 cancer using genomics and informatics. His lab has

6 made substantial contributions to open-access resources

7 for cancer research. Recently, the development of

8 bioinformatics for immunogenomics has become a major

9 focus of his lab.

10 Dr. Griffith now has more than 14 years of

11 experience in the field of genomics, bioinformatics,

12 datamining, and cancer research. He has over 80

13 publications and has received numerous research awards

14 and has held several large grants, including an NIH

15 K99. So I would like to welcome Dr. Griffith.

16 BIOINFORMATICS TOOLS FOR DEVELOPMENT, ANALYSIS &

17 PRECLININCAL TESTING OF INDIVIDUALIZED THERAPEUTICS -

18 DR. MALACHI GRIFFITH

19

20 DR. GRIFFITH: Okay. Thank you for inviting

21 me to speak today. It's been a really interesting

www.transcriptionetc.com

129

1 morning so far. I’m going to talk about something

2 that's been mentioned in several of the previous

3 presentations of bioinformatics but much more in

4 passing. So we're going to dive a little bit deeper

5 into the bioinformatics aspect of all this right now.

6 And I'd like to start by making an argument

7 that neoantigen vaccines in particular are a really

8 nice exemplar for individualized therapeutic. So we

9 heard a really interesting comment and question from

10 the audience this morning about neoantigen vaccines.

11 And I think that they're a great exemplar for a couple

12 of reasons.

13 One is that they're, in some ways, the most

14 personalized or individualized example of therapy that

15 we see today. So what's depicted here is a very high-

16 level 10,000-foot view of a workflow for developing a

17 neoantigen vaccine for an individual patient where you

18 start with a piece of their tumor tissue, taken at

19 biopsy or in some cases from a surgery. And then you

20 sequence the whole genome or whole exome of that tumor

21 DNA and compare it to the normal DNA from that

www.transcriptionetc.com

130

1 individual, from blood usually.

2 And you would usually also sequence the

3 transcriptome of that individual and then do a very

4 complicated bioinformatics analysis, which I'm going to

5 dive into a little bit more deeply here, to identifying

6 variation that leads to unique amino acid changes in

7 the genome of the tumor. So these are peptide

8 sequences that are specific to the tumor cells. And

9 then you use knowledge of those peptides on how the

10 immune system works to try to develop a vaccine made up

11 of these peptides that will stimulate the person's

12 immune system to respond to their tumor.

13 And so a typical example of this right now, in

14 many of the clinical trials that are underway, is

15 something like 5 to 20 peptides that are unique to the

16 tumor cells that you manufacture in a variety of ways.

17 And there are various manufacturing strategies being

18 used that get delivered, in the hopes of stimulating

19 the immune system to attack their tumor. And those

20 peptides are totally unique to that individual.

21 They'll never be used again. You're leveraging the

www.transcriptionetc.com

131

1 unique features of that tumor genome.

2 They're largely passenger mutations, so they

3 don't need to be functional per se. They just need to

4 be present in every tumor cell and cause an amino acid

5 change. And because they're passenger mutations and

6 they don't drive the biology of the cancer, every

7 person has a somewhat random set of them.

8 So I've been involved in probably now 50 to

9 100 vaccine designs. We've never used the same peptide

10 twice. It's always a different set for every single

11 patient.

12 So it's sort of the pinnacle of individual or

13 personalized therapy. And the other sort of large

14 reason, I think, that it's a good exemplar is just the

15 scale of the target patient population. Since somatic

16 mutation is a fundamental feature of virtually all

17 cancers, the potential application is potentially all

18 cancer patients. Everyone has an adaptive immune

19 system, and everyone's tumor has unique features of

20 those cells that could be targeted by their adaptive

21 immune system.

www.transcriptionetc.com

132

1 So the number of people that could potentially

2 benefit from this strategy is huge. So you have a

3 combination of a challenge, which is that it's

4 completely personalized. There's never going to be an

5 off-the-shelf drug that you can give to someone. But

6 at the same time, the potential market is absolutely

7 massive.

8 So just to dive in a little bit more detail in

9 terms of what this pipeline looks like. So I don't

10 expect you to read this. It's just to -- along with

11 several slides, to give you kind of a sense of the

12 complexity and detail that goes into this.

13 So it all starts with sequence data. So we've

14 heard this morning how important access, robust access

15 to that data is. And there's a series of complicated

16 bioinformatic steps that are undergone to sort of

17 convert that in raw genomic information into

18 predictions of first variation.

19 So you align your sequences against a

20 reference genome, and then you perform a variety of

21 different kinds of genomic variant calling and HLA

www.transcriptionetc.com

133

1 typing to characterize the immune system of that

2 patient. And then you run those variations through

3 customized pipelines that attempt to prioritize and

4 rank these variants according to their potential

5 usefulness as an immunogenic target. So all of this is

6 incredibly complicated. And there are a ton of tools

7 out there. And it's becoming increasingly sort of

8 automated and robust. But it's still also quite an art

9 form.

10 So currently, the vaccine design process

11 involves considering quite a large number of factors.

12 And this is often done in an immunotherapy tumor board

13 setting for the trials that are experimenting with this

14 therapeutic modality. And this is a group of experts

15 in immunology, genomics, bioinformatics, and the

16 treatment of the type of cancer that's being targeted,

17 who consider a variety of criteria for each of the

18 candidates that you're thinking of making the

19 individual personalized vaccine for.

20 And these are some of the things that are

21 features of the patient, and some of them are of their

www.transcriptionetc.com

134

1 genome. And some of them are individual to the

2 particular variants that you're thinking of targeting.

3 And I've just listed some of the examples on the right

4 side. And really, what we're doing in my group is

5 trying to automate as much of this as possible,

6 formalize the stuff that still involves human

7 intervention, replace as much of the human interaction

8 with machine learning as possible, and develop sort of

9 best practices and SOPs to help make this process

10 really reproduceable.

11 So we've heard a lot this morning about

12 process. This is a great example of that. So the

13 therapy is always different, but the process really,

14 really matters. And it's an incredibly complicated

15 process. So it's right for people thinking deeply

16 about the details of it and how we can make it robust

17 and reproduceable and make the process accessible to as

18 many patients as possible.

19 So part of the reason why this is a moving

20 target and we're still developing new approaches is

21 that this is a very new treatment strategy. And we're

www.transcriptionetc.com

135

1 just still kind of learning the rules of what makes a

2 good immunogenic neoantigen, so the peptides that are

3 specific to a tumor that actually lend themselves to

4 this approach. And one of the best ways that we're

5 learning this is by doing it in early-stage trials in

6 patients. And I've listed the trials that I'm involved

7 in here, which are a relatively small set of the trials

8 that are underway worldwide.

9 As you can see, they involve a variety of

10 cancer types, and they’re relatively small scale being

11 mostly phase one trials. But they all follow the same

12 process, which is that we start with a patient sample,

13 a piece of their tumor. We sequence the genome of it,

14 and we go through a pipeline that is both bioinformatic

15 and manufacturing and a preclinical assessment to

16 arrive at a vaccine that is actually safe and able to

17 be delivered to the patient by injection either

18 peptides or a dendritic cell vaccine or, in some cases,

19 the vaccine is incorporated into vectors that are sort

20 of similar to some of the vectors that have been

21 described this morning for delivering genetic payloads.

www.transcriptionetc.com

136

1 There's a huge amount of variability in these

2 trials. So they vary in their delivery approach. As I

3 mentioned, there's several manufacturing and delivery

4 strategies being used, the site of the tumors that are

5 being considered. It's almost every cancer type now

6 that has one of these trials underway somewhere. These

7 are some of the examples of the ones that are happening

8 locally at Wash U.

9 And then they vary in their combination with

10 other therapies. So many of them are being combined

11 with checkpoint blockade therapy, where the idea here

12 is to sort of take the brakes off the immune system,

13 while also simultaneously giving a kind of roadmap to

14 the immune system, so telling the immune system, these

15 are the unique features of the tumor in this patient

16 that you should be targeting, and then simultaneously

17 sort of stimulating the immune system to attack. So in

18 many of the talks we heard this morning, there was

19 concern about sort of immune toxicity or the effects of

20 the immune system are almost a problem.

21 But here, it's the reverse. We're trying to

www.transcriptionetc.com

137

1 enrage the immune system against the tumor but also

2 sort of provide some guidance in terms of what

3 specifically it attacks. And the level of specificity

4 here is quite exquisite because it's this very

5 personalized genomic base therapy individualized to

6 each patient.

7 So what I'm going to do now is just walk

8 through a list of some of the bioinformatics concepts.

9 So these are the things that keep me up at night, when

10 we're thinking about how do we do this whole process

11 from raw data to vaccine design and production in a

12 reproduceable and robust way so that the process is

13 constrained, and given the same set of input data, you

14 would arrive at the same answer. And I'm not going to

15 list those here because I'm going to go through them

16 one by one.

17 So it all starts with the sequence data, which

18 must ideally be of high quality. This part of the

19 process has become quite robust and sort of production-

20 ified, if you will, where there are many, many

21 sequencing cores and facilities and services where you

www.transcriptionetc.com

138

1 can send nucleic acid for the tumor and for the

2 reference blood and get DNA and RNA isolated, libraries

3 constructed, sequences generated. And then you think

4 of this sort of commodity sequencing, where everyone

5 has sequencing from the same platform, and they kind of

6 all look the same. The reads will be a certain length,

7 and they'll be paired and so forth. Those are certain

8 standards that have been widely adopted.

9 And I think that's largely true. But I would

10 just caution at this stage that the apparent

11 consolidation of many sequencing efforts to single

12 sequencing platform can lead to a false sense of

13 reliability in terms of sequence quality and the nature

14 of each sequence dataset. So I think that assessing

15 quality of your raw sequence data is still very, very

16 important. But I won't go into much more detail on

17 that.

18 The analysis of the data really starts with a

19 reference genome. So people think of the human

20 reference genome project as complete. It's not really

21 complete. It's still ongoing. But there is this sort

www.transcriptionetc.com

139

1 of pervasive fallacy that there's one human reference

2 genome, which for a bioinformatician is very much not

3 true.

4 So while the raw assembly is

5 centrally maintained and you can sort of go and get the

6 centrally maintained build or assembly, there are many,

7 many, many derivatives of it. And these are in common,

8 common use. So we all hopefully know that there are

9 multiple assemblies or builds in common use. So we

10 think of build 37, build 38, or HG19, HG20. But there

11 are also many subversions, patches within those. And

12 the actual raw files used in a referenced genome can

13 matter.

14 And so usually, what's happening is people

15 aren't getting the raw assembly files themselves.

16 They're getting them through some second party, like

17 Ensembl, UCSC, 1000 Genomes, the Genome Data Commons.

18 And each of these vary in a variety of ways that can

19 really matter for downstream analysis and

20 interpretation. So for example, some of them used so-

21 called decoy sequences. Some of them include

www.transcriptionetc.com

140

1 alternative haplotypes and some don't.

2 There are light versions that simplify the

3 genome down to the chromosomes and throw away all of

4 the unplaced contigs. naming amazingly is

5 not consistent to this day. Some viral genomes are

6 included, and some referenced genome sequences repeat,

7 masking may vary, et cetera.

8 So this still remains a large problem in terms

9 of consistency. The Global Alliance is really working

10 on trying to standardize some of this by developing

11 SOPs and best practices for uniquely identifying what

12 your reference genome really is. Variant discovery is

13 another area of a lot of variability. So you start

14 with alignment. And then with an alignment, you run a

15 series of variant calling algorithms or transcriptome

16 analysis.

17 And there is a huge diversity of how these

18 algorithms work and how they're used in combination.

19 And there are generally a series of tools used for each

20 broad type of variation. So for example, you might

21 have three variant callers, strelka, mutect, and

www.transcriptionetc.com

141

1 varscan, just to pick some examples, for calling single

2 nucleotide variants in a different set of variant

3 callers for SVs. And so you could have dozens of tools

4 potentially involve, just in identifying the potential

5 variation that's the grist for the mill, to identify

6 your neoantigens for these personalized vaccines.

7 Because of the complexity of this process,

8 again, manual review human intervention remains a

9 common part of many of these pipelines, where humans

10 actually look at raw sequence data. They manually

11 review variant calls that they're going to invest a lot

12 of effort or make something around. And so that part

13 of the process can be variable. We've worked to

14 develop standard operating procedures for the manual

15 review aspect of this and, also, machine-learning tools

16 to help automate it so to take humans out of the

17 equation.

18 Once you have variance, so you have identified

19 a genome variation that could be a source of a

20 neoantigen, there's a big representation problem in the

21 field. We can't agree on how to refer to variance.

www.transcriptionetc.com

142

1 And this creates a lot of confusion and inconsistency

2 across the field.

3 So I'm showing at the top here six different

4 depictions of the same variants, all being named in

5 different ways, only one of which is actually

6 unambiguous and computationally interpretable, and even

7 then is not in a very efficient computationally

8 interpretable form. So that's the bottom one. And the

9 bullet point is an HGVS string. The others are just

10 essentially colloquial ways of referring to a variant -

11 - that many of us know what BRAF V600E means. But

12 that's a sort of ambiguous way of representing it, to

13 just name it like that.

14 So this creates a problem because it's

15 difficult to know when two resources or two groups or

16 two labs are talking about the same or different

17 variation, which creates a sort of consistency

18 challenge. Again, there are many efforts to harmonize

19 variant identification. I'm involved in several of

20 these consortia that are sort of having conversations

21 around how we develop standards and ontologies to

www.transcriptionetc.com

143

1 really fix this part of the problem.

2 And the one that I really like to mention

3 specifically is the ClinGen Allele Registry, which is

4 depicted on the right here. And this is not the

5 perfect solution, but it's available now. And it fixes

6 a lot of the variant identification problems for many

7 people.

8 Just like the reference genome has many

9 versions, the reference transcriptome has many

10 competing versions. And when you're trying to think of

11 an amino acid change that arises from a change in the

12 tumor genome, that relies on interpretation in the

13 context of a specific transcript sequence. And we

14 don't actually know what that is. It's an

15 interpretation. And so it relies on predictions for

16 what transcripts look like.

17 And there are many competing reference

18 transcriptome efforts. Each of them handle prominence

19 and versioning differently, which creates a problem for

20 building robust reproduceable pipelines. For example,

21 Ensembl versions each individual transcript and their

www.transcriptionetc.com

144

1 overall set. But they're currently on release 99,

2 which creates another problem, which is that it's

3 constantly shifting targets.

4 You always have this temptation to be on the

5 latest set of reference transcripts, but at the same

6 time, that creates an instability in your pipeline

7 where it makes it harder to kind of lock down the

8 pipeline, where you can run it multiple times and get

9 the same answer. But Ensembl is popular among

10 bioinformaticians because of their sort of formal

11 handling of certain aspects of this problem. But it's

12 really important.

13 So we've identified several examples in our

14 own recent experience in these clinical trials where,

15 for a given referenced transcriptome, how you interpret

16 genomic events can really radically change based on how

17 you prioritize transcripts and consequences. So

18 there's this one variant, one consequence problem. We

19 like to think of one variation in the genome leading to

20 one amino acid change in one gene. But it's more

21 complicated than that because of overlapping genes,

www.transcriptionetc.com

145

1 because of alternative isoforms of genes.

2 And so there's very complicated tools that

3 attempt to solve this problem for you. And they have

4 to make difficult choices about how to prioritize the

5 referenced transcript that you're using for your

6 inference. And this can create a variety of problems.

7 So two examples of problems we had recently where cases

8 where the variant effect predictor we were using

9 changed the way they internally represented frame

10 shifts. And this caused us to have the potential to

11 create incorrect peptide sequences. And we can also

12 miss high-priority targets when an inappropriate

13 transcript gets selected as the highest priority.

14 Clinical variant and gene interpretation is

15 another challenge that several consortia are now

16 working to resolve. So the -- in the sort of rare

17 human disease space, the ACMG guidelines have really

18 helped to solidify how we do variant interpretation.

19 In cancer, there's been -- we're sort of behind because

20 we have the somatic variant problem that's been less

21 addressed. But we're starting to make progress with

www.transcriptionetc.com

146

1 the AMP, ASCO, CAP guidelines and other efforts like

2 CVC. And again, the Global Alliance has really been

3 organizing efforts to improve the consistency of both

4 gene and variant interpretation in a clinical context.

5 And then I really just want to end on the sort

6 of most bioinformatics heavy aspect of this whole

7 process, which is that you have this incredibly

8 complicated analysis pipeline for individualized

9 therapeutics. I'm depicting a small slice of our

10 pipeline here as a graph, with nodes and edges,

11 depicting steps, tools, data coming in, and

12 interpretations coming out. That's actually a small

13 piece of the overall neoantigen vaccine prediction

14 pipeline that we use that goes from raw data to a new

15 antigen vaccine. It involves dozens of tools, hundreds

16 of parameter settings, hundreds of input and output

17 files, and thousands of individual compute steps, which

18 makes it very difficult to have an actual reproduceable

19 pipeline.

20 So it gives us this question of how do we

21 actually ensure reproduceable results for very complex

www.transcriptionetc.com

147

1 pipelines? And so this is an area that we've really

2 been working on formalizing. And the short answer is

3 you should adopt a formal way of describing your

4 pipeline first of all. So there's been several recent

5 advances in things like workflow definition languages.

6 You should containerize everything. So place

7 all of tools inside of containers that isolate the

8 environment from compute dependencies. And you should

9 use a workflow execution system that runs the whole

10 pipeline.

11 And then ideally, you should organize these

12 layers into an analysis platform. And there are some

13 great examples out there like TARA. And don't forget

14 the importance of software engineering and

15 bioinformatic support.

16 So it's harder than it seems to keep a

17 computational pipeline locked down. There's an

18 assumption among many that because a bioinformatics

19 pipeline is computational, it must be inherently stable

20 or reproduceable, and this is actually surprisingly not

21 true.

www.transcriptionetc.com

148

1 It's potentially stable and reproduceable.

2 But that's actually much harder to achieve than you

3 imagine because of the complexity and the number of

4 steps and the dependencies on environment and how much

5 those environments and inputs and reference files --

6 everything can change. And it's harder than it sounds

7 to keep things locked down. So I'd just sort of urge

8 caution when thinking about the reproducibility of

9 these pipelines.

10 And then I'll just end with a short list of --

11 so I've thrown a lot of terminology and resources and

12 efforts out, but this is sort of the short list of

13 things, that if you're just going to check out a few

14 things or you want to become engaged in this kind of

15 work, these are the four things that I would recommend

16 starting with. I'd be curious to hear if anyone has

17 any examples of bioinformatics issues for

18 individualized therapeutics that I really missed. And

19 then I'll just end by acknowledging the wonderful group

20 that I'm privileged to codirect with my twin brother

21 and partner in crime, Obi, and of course, my funding

www.transcriptionetc.com

149

1 from the NHGRI and NCI in incredibly grateful for. I

2 look forward to talking to you later if you have

3 questions.

4 DR. SANDUJA: Thank you, Dr. Griffith. So

5 we'll now move on to our next speaker, Dr. Keith Joung.

6 Dr. Joung is currently a Desmond and Ann Heathwood

7 Research scholar and pathologist at Mass General

8 Hospital and a professor of pathology at Harvard

9 Medical School. Dr. Joung holds a PhD degree in

10 genetics from Harvard University and an MD from Harvard

11 Medical School. He's a leading innovator in the field

12 of gene editing.

13 Dr. Joung has pioneered development of

14 important technologies for targeted genome editing and

15 epigenetic editing of human cells. He has received

16 numerous awards, including an NIH Director's Pioneer

17 Award, an NIH Director's Transformative Research

18 Project R01 Award, the MGH Research Scholar Award, and

19 an NIH R35 MIRA Award. So we would like to welcome Dr.

20 Joung.

21

www.transcriptionetc.com

150

1 DEFINING OFF-TARGET EFFECTS OF GENE EDITING

2 TECHNOLOGIES - DR. J. KEITH JOUNG

3

4 DR. JOUNG: Thank you. And thank you to Dr.

5 Marks and the other FDA staff for the opportunity to

6 come and speak. So I'm going to talk today about

7 defining off-target mutations and effects of gene

8 editing technologies broadly. I have a conflict of

9 interest slide, which I'm required to show you by my

10 institution.

11 And so what I hope to do today is three

12 things. One is review the challenges and strategies

13 that exist now for defining gene editing nuclease, as

14 well as sort of next-generation CRISPR-based editor

15 off-target effects, and then tell you about our latest

16 assay, which we call ONE-Seq, which is a universal

17 platform for identifying off-target effects of all gene

18 editing nucleases and CRISPR-based editors that we

19 think also has advantages of scalability as well as

20 reproducibility, and then, at the end, just share some

21 perspectives on kind of the state of the field and then

www.transcriptionetc.com

151

1 what I see as issues and challenges moving forward. So

2 first, starting with a review of the strategies and

3 challenges involved in defining off-target effects of

4 gene editing nucleases.

5 So as Dr. Marks mentioned in his intro remarks

6 this morning, there are now a variety of different

7 platforms for doing gene editing. There are the four

8 sort of classical, if you will, nuclease platforms,

9 zinc finger nucleases, TALENs, mega-nucleases, and

10 CRISPR CAS RNA-guided nucleases, and then more next-

11 generation technologies, such as the CRISPR-based

12 cytosine- and adenine-based editors developed by David

13 Liu's group, which used the CRISPR system to direct

14 specific nucleobase deaminase to specific

15 locations in the genome.

16 So we've gotten very good now at being able to

17 put mutations where we want in the genome. But one of

18 the big challenges for the field for many years has

19 been defining and quantifying where else in the genome

20 we may be making alterations other than our intended

21 on-target site. And part of the challenge is that the

www.transcriptionetc.com

152

1 alterations in use by nucleases and base editors are

2 double-strand breaks and nicks. And so these are

3 short-lived events that are then repaired by cellular

4 DNA repair processes.

5 And so it's hard to actually directly identify

6 these alterations. And instead, what we do is identify

7 their outcome, so things like nonhomologous mediated --

8 nonhomologous end joining mediated indels or the base

9 substitutions induced by base editors. This can be

10 challenging to do, particularly in repetitive genomic

11 regions. And because of limitations in our ability to

12 do sequencing, it can be hard to distinguish these

13 alterations relative to background mutations or just

14 errors in the process of doing the analysis itself.

15 And this is particularly true for base editors.

16 Another challenge is that there really is no

17 gold standard for the field for off-target

18 determination. Whole genome sequencing is neither

19 practical nor particularly sensitive for finding these

20 alterations. And then on top of that, because of

21 tremendous interest in the field, it's a rapidly

www.transcriptionetc.com

153

1 evolving space with continuous improvements and

2 discoveries coming at a very rapid pace.

3 So how does the field address this? Well,

4 essentially right now, we use a consensus two-step

5 approach for being able to identify gene editor off-

6 target mutations. So the first step is what's called a

7 nomination or discovery process. And here, what you're

8 trying to do is identify potential sites of off-target

9 cleavage or mutations in a surrogate setting.

10 And so that surrogate setting can either be

11 cell-based assays, like the GUIDE-Seq assay developed

12 from my group and bless/bliss methods developed by Feng

13 Zhang’s group. Or it can be in vitro methods, where

14 you in a test tube have purified genomic DNA and

15 purified nuclease or base editors, and you ask whether

16 you can identify off-target effects. Now, you want

17 this to be as sensitive a method as possible. And you

18 want it obviously to be genome-wide in scope, so that

19 you can define the superset, if you will, of all

20 possible sites that the editor might be making

21 alterations at.

www.transcriptionetc.com

154

1 It's also important to note that not all sites

2 identified in this step may ultimately be -- ultimately

3 show evidence of alterations in the context of the

4 cells that you actually want to modify for a

5 therapeutic because there are other factors that come

6 into play that would be specific to your therapeutic

7 setting, such as, for example, the epigenetic status of

8 the gene. So the second step then is to take all of

9 the sites that you get from the nomination process, and

10 then ideally in the setting, therapeutic setting of the

11 cells that you want to be able to the therapy in,

12 actually look directly at those sites and ask whether

13 or not you see evidence of alterations at those sites.

14 And so this is why it's really important for that first

15 step to be as sensitive as possible because, if you

16 don't identify it in that first step, you won't even

17 look at it in the second step.

18 And here, there are a number of challenges,

19 which I'm not going to read through, but that also

20 exist with identifying whether an alteration has

21 occurred here. Typically, what people have been using

www.transcriptionetc.com

155

1 is targeted amplicon sequencing to be able to look for

2 the presence of indels. But it is important to note

3 that it is equally important to try to identify other

4 types of alterations that can occur when you create

5 more than one double-strand break in the cells, so

6 things like large-scale inversions or deletions or

7 translocations.

8 And then overall, I think it's important to

9 try to quantify risk as the sum, if you will, of the

10 on-target effect, but also all of the off-target

11 effects and the overall double-strand break burden that

12 you're actually inducing in the cell. Now, there's a

13 number of critical parameters. I don't have time to go

14 through all of them in a talk this short. But I do

15 want to mention assay sensitivity as being one that's

16 particularly important.

17 As I mentioned, for the nomination or

18 discovery step, this is very important because, if

19 you're not sensitive enough to pick up everything,

20 again, you won't even bother to look at it in the

21 second confirmation step. And in vitro assays, I

www.transcriptionetc.com

156

1 think, have the advantage over cell-based assays for

2 the nomination step of being more sensitive. We can't

3 quantify that sensitivity at this point, again, because

4 of the low frequency sites having some challenges being

5 able to confirm due to the error rate of next-gen

6 sequencing, which also limits the second confirmation

7 validation step as well.

8 Because if you amplify a section segment of

9 the genome, the process of doing PCR and then doing

10 next-generation sequencing, you can often see indels at

11 those sites at rates anywhere from 0.1 to 0.01 percent.

12 And so distinguishing a real alteration introduced by

13 your base editor or nuclease of interest from just

14 background error rates can be challenging. And then

15 ultimately, too, risk assessment is important because

16 you may make a break, but it may not have any --

17 ultimately any functional consequence. But here, we're

18 limited to some degree, or to a large degree, by our

19 understanding or knowledge of biology and genome

20 function at a particular identified off-target site.

21 Assay quality control is also equally

www.transcriptionetc.com

157

1 important. So some of the parameters to think about

2 here are positive and negative controls, particularly

3 for a negative study that shows or reports no

4 detectable off-target effects. It's also important to

5 account for sequence variation relative to reference

6 genome sequence.

7 And we've just heard about some of the

8 challenges, even in what constitutes referenced genome

9 sequencing in and of itself. But certainly,

10 differences among different cell types are important to

11 account for, so doing an untreated control is very,

12 very important when you're looking for off-targets.

13 And then there's a lot of other parameters, which

14 again, I don't have time to get into, but that relate

15 to the number of input genomes that are going into your

16 assay, the number of assays that you do, and assay

17 replicates that you do and biological replicates that

18 you do, sequencing depth, and then all the informatic

19 pipelines that you use to actually process the data.

20 So there are a lot of challenges. The NIST

21 has formed a consortium on gene editing led by Samantha

www.transcriptionetc.com

158

1 Maragh that I think is trying to address some of these

2 challenges. And it may be that, ultimately, commercial

3 service providers may be able to help with providing a

4 standardized trusted set of services that would address

5 these requirements.

6 Okay. So next, I want to tell you about a new

7 assay that has been recently developed by my lab. And

8 again, this is unpublished work. And here, it's an

9 assay we call ONE-Seq, which we think provides -- we

10 believe provides a universal platform for being able to

11 identify off-target mutations of gene editors both of

12 the nuclease class and the base editor class and to do

13 so with unsurpassed sensitivity.

14 So this is an in vitro assay, again, purified

15 components, in a test tube. If you look at all the

16 other assays that have been described previously in

17 this field, so things like Digenome-seq, SITE-seq, and

18 even CIRCLE-seq previously described by my lab, what

19 those assays do is they purify genomic DNA out of cells

20 or a particular tissue. And then they build some kind

21 of library out of that, treat with the nuclease, and

www.transcriptionetc.com

159

1 then attempt to identify in the context of all these

2 genomic sites, which are the sites that are actually

3 being cleaved by the nuclease.

4 And so the representation in the middle is

5 sort of the mess of genomic DNA, if you will, that you

6 get. In the human genome, obviously, you're going to

7 have at least three times 10 to the 9 of different

8 sites. And only a very small number of these will

9 actually end up being cleaved by the nuclease of

10 interest.

11 And what we've learned from a variety of

12 different studies performed to date is that these sites

13 all have some degree of resemblance to the intended on-

14 target site of the nuclease or the base editor. That

15 is that they are the same, but they differ at a certain

16 number of positions, as many as six or seven, within

17 the target site that you're trying to hit. And so it's

18 important then to emphasize that this is a very, very

19 small number of sites relative to the total content, if

20 you will, in a genome sequence.

21 So with ONE-Seq, what we do is we take whole

www.transcriptionetc.com

160

1 genome sequence for a particular cell type -- so for

2 example, for a human genome, we may use human reference

3 genome sequence -- and we go through and

4 computationally identify all the sites that have a

5 certain number of mismatches relative to the on-target

6 site. So you can go through and computationally

7 identify these. And you end up, for example, with

8 Cas9, typically, you have a list of sites of anywhere

9 from about 20,000 to 80,000 sites, depending on the

10 degree of orthogonality relative to the human genome

11 sequence.

12 And so you can then extract these sites out of

13 the genome and then synthesize all of them using high

14 throughput oligonucleotides synthesis, which now has

15 the capability to be able to synthesize up to millions

16 of these sequences precisely and give you exactly what

17 you want. And so what we do is we embed these

18 sequences in a fixed-length oligonucleotide. They're

19 always at the same position in the middle. And all of

20 these oligos are the same length.

21 And then after synthesis, they can be released

www.transcriptionetc.com

161

1 from the chip, converted to double-strand DNA. And

2 then this becomes your library that you then treat with

3 your nuclease of interest. And then I'll show in a

4 second how we extract the off-target cleavage sites.

5 So this type of approach using, if you will, a

6 more focused library that's been synthesized in vitro

7 relative to just using genomic DNA, has a number of

8 advantages. So one of them is that you can

9 characterize the entire library when you build it, just

10 by doing a simple MiSeq run because you're talking

11 about, again, anywhere from 20,000 to 80,000 sites.

12 And so you can sequence that library at high coverage

13 and know exactly what's going into the reaction in the

14 first place.

15 And so these are some examples of multiple

16 libraries that we've built for different target sites.

17 And you can see that the dropout of sites is actually

18 very, very low. It's also very, very reproduceable,

19 these libraries. So you do two independent syntheses,

20 and then you do sequencing and compare them. The

21 reproducibility of this is very, very high. And so

www.transcriptionetc.com

162

1 these are two different libraries here, which we've

2 done in duplicate and then done high-coverage

3 sequencing. And you can see that the reproducibility

4 between the libraries is also very high.

5 So this is a description of how we then use

6 this approach with Cas9 nuclease. So you take this

7 library, where you have a bunch of these different

8 target sites. You then treat with the Cas9 nuclease.

9 And so all the sites that are cleaved will be broken

10 into two. And then those free ends then serve as

11 for ligating a sequencing adapter. And then

12 you can sequence the products that come out of this and

13 know which sites are being cut.

14 We then process the data. And so you end up

15 with these types of outputs, where you identify a whole

16 bunch of different sites. And typically, the highest

17 site, although not always, is the on-target site, shown

18 here with an asterisk. And you can assign what we call

19 a ONE-Seq score, which represents quantification of how

20 frequently these sites are being cut in the in vitro

21 reaction.

www.transcriptionetc.com

163

1 Now, another nice thing about this assay is

2 the unique capability to be able to set false-positive

3 thresholds for the assay. This is something that you

4 can't really do with a genomic DNA library. And the

5 way that you do this is for a given target site

6 library, let's say against a target in the FANCF gene,

7 instead of treating that library with a FANCF-targeted

8 nuclease, you target it with a different nuclease

9 target it to a different gene, so for example, to an

10 EMX1 target site.

11 And so what that allows you to do then is see

12 what the false positive rate is because nothing in that

13 library should be cut by a nuclease that doesn't target

14 that site. And so you can set precise cutoffs. And so

15 you can see the scatter plot on the right here are two

16 different library experiments. And again, you see high

17 reproducibility in the ONE-Seq scores of the different

18 sites. And the red lines represent the false positive

19 cutoffs that can be set based on doing these types of

20 mismatched nuclease experiments.

21 Okay. So to show you that ONE-Seq performs at

www.transcriptionetc.com

164

1 least as well, if not actually better than all the

2 other previous existing methods, here are comparisons

3 for four different sites of ONE-Seq against our GUIDE-

4 Seq cell-based method. And this is for identifying

5 bona fide, verified cleavage sites that actually are

6 cut in human cells. And so you can see that GUIDE-Seq

7 identifies -- sorry, ONE-Seq identifies all of the

8 GUIDE-Seq sites but then also identifies additional

9 sites as well.

10 This is a comparison of how well CIRCLE-Seq,

11 another method we had previously described, an in vitro

12 method we previously described, performs at finding

13 these bona fide GUIDE-Seq sites. And you can see the

14 CIRCLE-Seq sometimes can miss the different sites. The

15 two different colors represent doing CIRCLE-Seq on two

16 different cell type DNAs. And then finally, Digenome-

17 Seq, which is another in vitro method previously

18 described by Jin-Soo Kim’s lab, you can see also

19 misses sites, bona fide GUIDE-Seq sites. Although,

20 this was a result that's sort of known from the

21 previous literature.

www.transcriptionetc.com

165

1 We wanted to show that ONE-Seq was actually

2 capable of identifying sites in an in vivo context as

3 well. And so here, we used a liver-humanized mouse

4 model. And so this is a mouse model, where

5 essentially, a good portion of the mouse liver has been

6 replaced by human hepatocytes.

7 And so the nice thing about this model is it

8 allows you to examine off-targets in the context of

9 human cells but in a mouse model. And so this

10 addresses something that actually Dr. Marks was talking

11 about this morning, that when you want to look at off-

12 targets in a mouse model that's not really relevant to

13 look at off-targets in a mouse genome if ultimately the

14 goal is to use these nucleases for human therapeutic.

15 So I'm not going to go into the details of how we build

16 these types of mice. And I should say that this is

17 work done with Karin Musunuru at UPenn in collaboration

18 with his group.

19 So we wanted to do a particularly challenging

20 site. So we -- Karin had identified this GUIDE RNA for

21 Cas9 that targets early exon in the PCSK9 gene. And

www.transcriptionetc.com

166

1 the nice thing about this GUIDE is that it has very few

2 closely matched sites in the genome. So it has no off

3 by one, off by two, or off by three sites. So the most

4 closely matched sites are off by four and then go up

5 from there.

6 And so we like this site because we thought it

7 would be particularly challenging to find off-targets

8 for this type of site, and we wanted to see how ONE-Seq

9 would do in identifying potential sites that ultimately

10 would be modified in vivo. And so this is the ONE-Seq

11 output. The top site here would be the on-target site.

12 And then underneath are the off-target sites. Little

13 colored squares indicate mismatches relative to the on-

14 target site. And on the left column, there are the

15 ONE-Seq scores.

16 And so these are the top 40 sites, off-target

17 sites, identified by ONE-Seq. Karin then went and took

18 genomic DNA from the livers of these mice that have

19 been treated with this nuclease and asked whether you

20 could identify indels at those sites. So you can see

21 on the right that, the on-target site, you get very

www.transcriptionetc.com

167

1 high modification. And then within this set of 40

2 sites, there are at least four sites where we can see

3 very reproduceable, in triplicate, evidence of indel

4 mutations.

5 And these are sites that have either four

6 mismatches, the two higher sites, or five mismatches

7 relative to the on-target site. And you can see from

8 the numbers of sites there that it would be very hard

9 if you went through -- wanted to go through and

10 actually look at every single one of these sites in

11 these livers. But here, we're able to sort of rank-

12 order the sites based on ONE-Seq and then focus on

13 those sites and quickly identify off-target.

14 So this validates that ONE-Seq is capable of

15 finding these sites in this in vivo context and in this

16 more therapeutically relevant mouse model system as

17 well. I don't have time to go through all the data,

18 but we have a lot of data showing that ONE-Seq

19 outperforms other methods for other types of nucleases.

20 So Cas9 is a nuclease that leaves a blunt end. But

21 there are other nucleases that leave overhangs, like

www.transcriptionetc.com

168

1 Cas12a (Cpf1) or engineered zinc finger nucleases, mega

2 nucleases, or TALENs.

3 And here's some data showing you that ONE-Seq

4 outperforms GUIDE-Seq for identifying Cas12a nucleases.

5 So we find all the sites previously identified by

6 GUIDE-Seq. But we also find additional bona fide sites

7 using ONE-Seq. And we've also adapted ONE-Seq for the

8 base editor technologies as well.

9 And so here, we show that for a variety of

10 different sites -- that for the sizing-base editors,

11 ONE-Seq outperforms the Digenome-Seq assay, which is

12 the only assay that's been used to date to identify

13 off-target sites for cytosine-based editors. And it

14 also outperforms Digenome-Seq for adenine-based editors

15 as well. So we believe that ONE-Seq is, as I say, at

16 least as good, if not actually superior to all the

17 other methods out there for identifying off-target

18 sites for nucleases that leave blunt ends, overhang

19 ends, as well as for the cytosine- and adenine-base

20 editors.

21 Okay. So I have a few minutes left because,

www.transcriptionetc.com

169

1 actually, I think the intro was counted in my 20

2 minutes. So I think I have a couple more minutes to

3 talk about briefly some perspectives on looking

4 forward. So first of all, some viewpoints and

5 perspectives on the current situation. So it is not

6 possible to really ensure a complete lack of off-target

7 effects at present.

8 And I don't think it's a reasonable goal to

9 ensure that, given where our technologies lie right

10 now. The goal really should be to minimize off-targets

11 as much as possible. And this can be done through

12 things like protein engineering and other technologies

13 that have been described in the literature over the

14 last few years -- but still understand that they may

15 occur. And also, there needs to be an understanding

16 that not all off-target mutations will necessarily be

17 problematic.

18 We also need to recognize that there are

19 sensitivity limitations of the existing assays and that

20 there's also restrictions imposed by sampling for both

21 ex vivo and in vivo therapeutics. You just can't look

www.transcriptionetc.com

170

1 at all of the cells that are actually being modified.

2 Off-target profiling, it's also important to remember

3 is only one aspect of safety analysis. So there are

4 other tox studies that still need to be done. And in

5 general, when you talk about risk assessment of off-

6 targets, it's not possible to really make a general

7 recommendation about specificity outside of the given

8 intended use of a particular nuclease.

9 So looking forward, I do want to say that my

10 own personal opinion is that it is very, very important

11 that we continue to try to improve and extend both the

12 experimental and the computational approaches for

13 identifying off-target effects. There are a growing

14 number of voices now in the gene editing space,

15 particularly on the kind of academic research side but

16 also to some degree from the industry side, that some

17 folks will say, well, this is -- we've done enough.

18 We've done enough to be able to look at this.

19 And so I would strongly disagree with those

20 opinions. I think not continuing to do so, given the

21 limitations and sensitivity that existing technologies

www.transcriptionetc.com

171

1 have, and does a disservice to patients because these -

2 - especially as these technologies become more widely

3 used beyond a small number of diseases. At the same

4 time, I want to emphasize that I think it's important

5 that we not get hung up on trying to get a situation

6 where we have nothing that is -- we believe that no

7 off-target effects are happening.

8 So it's important for the sake of patients to

9 be able to define risk as well as we can at any given

10 moment and then to balance that against benefit. And I

11 continue to be very bullish and very, very enthusiastic

12 about trying to move forward with these different gene

13 editing therapeutics into the clinic. We do need to

14 continue to improve sensitivity as well as to be able

15 to have assays that predict functional consequences of

16 off-target effects. And I do think, ultimately, a lot

17 of the limitations in sensitivity we have come down to

18 error rates of next-generation sequencing technology.

19 So providing strong support for the development or

20 advancement of next-gen sequencing technologies and

21 improving their error rates will be very, very

www.transcriptionetc.com

172

1 important for the future as well.

2 There's other newer technologies. So David

3 Liu's group recently described prime editors. And so

4 we are working on trying to actually adapt ONE-Seq for

5 prime editors as well. And then there are other types

6 of, if you will, Cas9-independent or non-sequence

7 recognition-based edits that can occur. So for

8 example, we recently described RNA off-target edits

9 that occur with the cytosine- and adenine-based editors

10 that are due to the deaminase portion of those enzymes

11 functioning on their own. And other groups have

12 described off-targets on DNA that are not -- of base

13 editors that are not necessarily guided again by the

14 Cas9. And so these are important areas to continue to

15 push forward on as well.

16 And then another very important area that I

17 believe will be important to account for in the future

18 is human genetic variation. So at the end of the day,

19 these are sequence-specific reagents that are using to

20 target sites. And so the profiles of off-targets are

21 not going to be the same in everyone. You can define

www.transcriptionetc.com

173

1 them for a reference genome, but it's actually very

2 important, ultimately, to be able to define them for

3 specific individuals if not actually specific

4 populations or subpopulations of individuals.

5 And although it's not possible to do this

6 practically or feasibly for every single patient at

7 present, I think advancing technologies that allow us

8 to better understand the impact of human genetic

9 variation on off-target profiles is a very, very

10 important goal for the future. And ultimately, being

11 able to define off-target profiles individually for

12 patients is important as well. And then it's also

13 important to continue to focus research efforts on

14 better functional assays that allow us to identify what

15 the functional consequences are and, in particular, for

16 tumorigenic risks of off-target effects because, again,

17 this is an area where we just don't have very good

18 assays at present. And then in the longer-term,

19 follow-up studies and considering how to -- how and how

20 long to look at patients going forward are important

21 areas of continued development and research as well.

www.transcriptionetc.com

174

1 So I will stop there. I'm only like a minute-

2 and-a-half, I think actually overtime. I do want to

3 acknowledge Vikram Pattanayak and Karl Petri in my

4 group, who led the development of the ONE-Seq assay.

5 And this was largely funded by DARPA, with some

6 additional funding from NIH and the MGH Research

7 scholars, and done in collaboration with the Lie Lab,

8 so David Liu's group at Harvard and Greg Newby.

9 And then I also want to acknowledge the

10 American Society of Gene & Cell Therapy, who had some

11 input in some of the slides that I did early on for

12 another presentation I gave about a year-and-a-half ago

13 as well on this topic. So thank you for your

14 attention. And I guess I'll be happy to take any

15 questions about these issues in the Q&A. Thanks.

16 DR. SANDUJA: We'll start with our panel

17 discussion now. I'd like to welcome our speakers and

18 Dr. Zuben Sauna from OTAT to join us. Yes, please.

19

20 PANEL DISCUSSION WITH Q&A

21

www.transcriptionetc.com

175

1

2 MS. HOWARD: Hello. I'm Marilyn Howard from

3 the University of Pennsylvania. I have a question for

4 Dr. Griffith. I was very intrigued by the multiple

5 steps that you have in your bioinformatics process.

6 And I'm wondering what the metrics are for feeding back

7 to the machine learning in that process and whether or

8 not any of the clinical trials have yet reached a stage

9 where the clinical outcomes can feed back into the

10 machine learning.

11 DR. GRIFFITH: That is a great question. The

12 short answer is that it's pretty early days for the

13 clinical trial, so none of those clinical trials have

14 actually completed yet. And many of the trials involve

15 vaccination in a setting where the tumor is not

16 actually on board anymore, so they've been -- the tumor

17 has been surgically removed. And the vaccine is being

18 used in almost like a vaccine sense to prevent

19 recurrence. So it will take some time for the survival

20 information to accumulate. So we're probably at least

21 two or three years away from that sort of real gold

www.transcriptionetc.com

176

1 standard of is this actually improving outcomes.

2 That being said, you mentioned the word

3 metrics, which I love. There's a lot of other earlier-

4 stage things we can look at. So many of the trials are

5 building in a variety of innovative immunological

6 monitoring steps to try to get an early sense of are we

7 seeing t-cell responses, in terms of changes on the TCR

8 repertoire or functional assays of t-cells, screening

9 for particular candidates before and after vaccination.

10 So that data should come sooner.

11 It's still going to be relatively sparse

12 because the trials are so small. But we can probably

13 learn quite a lot on a per peptide basis because many

14 of the assays are giving us a specific readout and

15 useful metrics for each of the peptides that we tested.

16 In some of these patients, we have up to 20 candidate

17 peptides. And we really don't know what the rules are

18 for what makes a good peptide.

19 So we're potentially going to learn a lot in

20 the next few years, subject to sufficient sharing of

21 that data and all of the usual challenges that go into

www.transcriptionetc.com

177

1 these kinds of studies. But I think that there's

2 reason to be very optimistic that we'll improve our

3 understanding of how to actually design a vaccine

4 significantly in the next few years and hopefully, more

5 like four to five years for the gold standard survival

6 analysis.

7 MS. HOWARD: Thank you.

8 MS. ADOMAKO: Hi. My name is Jessica Adomako,

9 and I'm from Genentech. This question is for you. And

10 it's --

11 DR. SANDUJA: Can you please speak up? It's

12 hard to --

13 MS. ADOMAKO: Can you hear me?

14 DR. SANDUJA: Yes.

15 MS. ADOMAKO: Yeah. This question is a follow

16 on to what the previous person asked. And there's a

17 second part, which is to the FDA. You talked a lot

18 about the bioinformatic challenges. And I completely

19 agree with you. And my question is have you looked to

20 the work that has already been done by the community at

21 large in establishing standards for analytic validity,

www.transcriptionetc.com

178

1 everything from referenced genomes to databases to

2 validity of bioinformatic pipeline software, et cetera,

3 et cetera, in the related field of developing NGS-based

4 tests?

5 Because there's a lot that can be learned.

6 And it's the exact same question to the CBER folks on

7 the panel. Your compatriots at CDRH have really done

8 an amazing job of establishing widely accepted

9 community standards.

10 And what we would like to know is are these

11 learnings being shared? Are there things that you can

12 develop based on what they've already established? And

13 completely conceding that this field is, as you said,

14 constantly evolving, we're always chasing a new goal.

15 But can we ping off of what we've already done?

16 DR. GRIFFITH: Yeah. So that's a really great

17 point. And I meant to mention this actually in my talk

18 that I think that there are many, many common themes

19 and similarities between what is going on, for example,

20 in the new antigen vaccine design process that can

21 benefit from the years and years of labs producing,

www.transcriptionetc.com

179

1 conducting genomic diagnostic tests and producing a

2 clinical report with interpretations for a specific

3 variance. They face many of the same challenges.

4 So we absolutely can learn a lot from those

5 practices and standards. And we're definitely -- all

6 of the consortium that I mentioned really evolved out

7 of those groups and heavily involve experts that have

8 years and years of experience in those areas. So yes,

9 absolutely.

10 MS. ADOMAKO: I’m just waiting to hear from

11 someone from CBER. Are you speaking here?

12 DR. SAUNA: I didn't quite get what do you

13 want -- what is the question to CBER?

14 MS. ADOMAKO: The question to CBER is, has all

15 of the work that's gone on at CDRH in establishing

16 these protocols, processes -- they even have wonderful

17 final guidance documents that are now being widely used

18 by sponsors. Is any of that, do you think,

19 translatable to CBER?

20 DR. SAUNA: I think Dr. Carolyn Wilson wants

21 to address your question.

www.transcriptionetc.com

180

1 DR. WILSON: I'm going to just step in real

2 quickly. Carolyn Wilson from Center for Biologics. I

3 just wanted to say, yes, we actually have at the agency

4 level a genomics working group, where we've actually

5 been exchanging and learning from other centers,

6 working in this space for about five years. And then

7 we also have a CBER genomic working group. And we've

8 actually brought the CDRH guidance documents to that

9 group and looked at them very carefully, as well as ICH

10 guidance documents and so on. So certainly, we're

11 well-aware of those other efforts and are incorporating

12 them into our own thinking.

13 MS. ADOMAKO: Thank you.

14 MS. WITKOWSKY: Hi. Lea Witkowsky from

15 Innovative Genomics Institute at UC Berkley and UCSF.

16 That was a really great session. Thank you, everybody.

17 And I have a question for Keith Joung in particular.

18 Very exciting data.

19 I'm wondering for ONE-Seq, if you're starting

20 with a prediction algorithm, presumably, you're

21 starting that -- you’re running that prediction on some

www.transcriptionetc.com

181

1 sort of reference genome. And as we think about

2 individualized therapies and kind of harkening back to

3 the vaccines, developing these at an individual level,

4 how do you expect, or do you expect, to be able to

5 adapt that for individual patients?

6 And you mentioned a little bit about this

7 human variability. And what is the capacity, or where

8 do you see the future going to be able to run something

9 like that to catch things that might be off-targets

10 only for an individual that happens to have a mutation

11 that makes it a new off-target that you wouldn't catch

12 normally in a reference genome, for example?

13 DR. JOUNG: Yeah. That's a great question.

14 Thanks, Lea, for that. So I think one of the strengths

15 of ONE-Seq is the ability to be able to look in a

16 detailed way at specific sequence changes for an

17 individual, or potentially even a group of individuals.

18 It is dependent on having all genome sequence

19 data for that person. When we set out to start

20 developing ONE-Seq actually about two, two-and-a-half

21 years ago, we assumed two things would start to come

www.transcriptionetc.com

182

1 down in cost. So one would be oligonucleotide

2 synthesis, which I think has already begun to play out,

3 and hopefully will continue to only get less and less

4 expensive. And the other was the assumption in the

5 continuing drop in the cost of being able to do whole

6 genome sequencing.

7 So if you envision a world going forward where

8 it's relatively inexpensive to do whole genome

9 sequencing and to do oligosynthesis, then I think it

10 becomes very reasonable to assume that you could

11 combine those two, be able to practice ONE-Seq, and

12 then be able to get information that's specific to a

13 particular individual. I also think it's just

14 generally easier to scale something like ONE-Seq to

15 cover more people and variants in more people than it

16 is to scale something like doing one-off in vitro

17 assays, the way you do Digenome-Seq, CIRCLE-Seq, or

18 SITE-Seq right now.

19 So I hope that makes sense. And that is our

20 hope, is that we will be able to account for more

21 individualized genetic variability because, as I said,

www.transcriptionetc.com

183

1 at the end of the day, these things are sequence-

2 specific agents. And so they are -- you expect -- it's

3 entirely reasonable to expect that their impact will be

4 specific to a particular individual based on their

5 genome sequence.

6 MS. WITKOWSKY: Thanks.

7 MR. STEIN: My name is Aron Stein with Sangamo

8 Therapeutics. This is a question for Dr. Joung. This

9 is in regard to your methodology for the validation of

10 your targets using a humanized mouse. Why that model

11 versus primary human hepatocytes?

12 DR. JOUNG: You could do it certainly in

13 primary human hepatocytes as well. We validated ONE-

14 Seq-predicted sites in cells in culture. So there's no

15 reason why you couldn't do it that way. Although there

16 are some challenges with getting the reagents

17 efficiently into human hepatocytes. It's certainly not

18 unreasonable to try that experiment.

19 The reason we did it in the context to the

20 mouse was that we wanted to look at in vivo in an in

21 vivo setting, where you would be delivering these

www.transcriptionetc.com

184

1 reagents in an in vivo setting, whether we would

2 predict those off-targets there. Because if you look

3 at the literature, to date, no one has actually been

4 able to identify these off-target sites, especially

5 those that have a large number of mismatches relative

6 to the on-target site in the context of something like

7 an in vivo animal model. In fact, our -- especially

8 when you're using a nuclease that has been designed to

9 be relatively orthogonal to the human genome. And so

10 that's why we chose that particular guide because it

11 would be easy to spot an off by one, off by two, off by

12 three. That I don't think anybody would be surprised

13 by.

14 But this is the first demonstration to our

15 knowledge where you're able to find off by fours or off

16 by five sites in an in vivo animal model where you've

17 delivered the nucleases in that way. So there were a

18 number of reasons for doing it. You certainly could do

19 it in cells. I didn't mean to imply that you couldn't

20 do it that way.

21 DR. SEYMOUR: Okay. Thank you.

www.transcriptionetc.com

185

1 MR. ALDRICH: Time for one more? Okay. I

2 just wanted to follow up with Malachi on his answer to

3 a previous question. Regarding -- I think a lot of

4 folks don't quite appreciate that the biggest problem

5 that comes out of the predictive algorithm is false

6 positives for neoantigens that aren't really present or

7 -- and that that's a problem which we can address by

8 running spot tests against -- ELISpot spot tests

9 against t-cells and eliminating false positives by

10 identifying, of the predicted and synthesized top

11 candidate antigens, synthesizing the peptides that

12 correspond to the neoantigens and then testing them

13 against the patient's t-cells for reactivity.

14 We kind of eliminate that problem of the false

15 positives in the -- and in terms of issues that are

16 really front and center for me as a patient, it's

17 making sure that, of the 5 to 20 peptides that we're

18 going to use in a final vaccine, that they're all

19 validated in some sense as having a corresponding T-

20 cell, which, hopefully, they'll amplify. I just

21 wondered if you'd comment on that. Thanks.

www.transcriptionetc.com

186

1 DR. GRIFFITH: Yeah. I mean it's a really

2 interesting question and topic that I think both the

3 false positives and false negatives are interesting. I

4 think that we think of the false positives as being

5 more tractable because as you say, you're right. We

6 can think about validating them further, and we can

7 look for specific T-cell responses.

8 Although, I guess I would say that it's still

9 falling short of -- ideally, we would know that they

10 were not just T-cell immunogenic but therapeutically

11 useful. And that's sort of like the next stage of --

12 MR. ALDRICH: Yeah.

13 DR. GRIFFITH: -- another layer of false

14 positives that's yet to be learned about.

15 MR. ALDRICH: Right.

16 DR. GRIFFITH: But I think we also don't know

17 what we don't know, in terms of false negatives. So we

18 don't really have a great sense, of the candidates that

19 we're nominating, how many great candidates did we

20 leave on the table just for not knowing about them, not

21 looking for the right kinds of variation or

www.transcriptionetc.com

187

1 prioritizing them incorrectly because of our lack of

2 complete understanding of how the immune system works.

3 So I think that's also an area for significant

4 improvement. And we are starting to see a little bit

5 more by unbiased assays looking at with the peptide

6 mass spec elution dataset started to give you a bit of

7 a sense of just sort of serving, like what are all the

8 peptides that we're sticking to a particular MHC

9 molecule and getting a more comprehensive readout of

10 that. Although, it also has some pretty significant

11 caveats to those datasets that we could probably talk

12 about for an hour.

13 MR. ALDRICH: For -- we will. But just one

14 last thing to have you comment on is one of the things

15 that ties into the talk we heard earlier, about

16 platform versus product and characteristics of the

17 neoantigen platform, as opposed to the specific vaccine

18 -- one of the things that I'm very impressed by is

19 that, when you read across the literature, we have a

20 great deal of familiarity with peptide vaccines. I

21 mean, historically, it's something where there's a lot

www.transcriptionetc.com

188

1 of data, not necessarily neoantigen peptide vaccines,

2 but peptide therapies have been around a long time.

3 And I think that one of the problems or one of

4 the challenges is that we have to recognize when we

5 have a platform where we know enough, so that we can

6 declare the platform as relatively safe. And as a

7 consequence, if you perform best practices, with

8 respect to the neoantigen platform, you can be pretty

9 well-assured that a patient isn't going to drop dead

10 when they're -- in fact, one of the incredible things

11 about the neoantigen trials that are going on is that,

12 to my knowledge, there hasn't been a single severe

13 adverse event reported from a safety point of view.

14 And yet, we are still waiting for the first

15 approval of a neoantigen peptide vaccine for any

16 indication of cancer. And so one of the things that

17 occurs to me is that, gee, if we have a safe platform

18 and we know enough about it and it proves efficacious

19 in phase-one trials saving lives, what is -- where do

20 we get comfortable enough to say, oh, well, if you're

21 following this best practice in terms of the supply

www.transcriptionetc.com

189

1 chain, we really ought to make it available to lots of

2 cancer patients? Just that's where I'm coming from.

3 DR. GRIFFITH: Yeah. I mean I guess my main

4 comment would just be to completely agree with you. I

5 think the pieces are in place to create such a platform

6 or process that we can really carefully document and

7 become confident that it is robust and reproduceable

8 and safe. My impression is -- as I said, many of these

9 have not published. Basically, none of them have. But

10 the early impression does seem to be that the safety

11 profile of this approach is outstanding.

12 And so that comes back to my initial comment

13 that the potential patient population this could be

14 applied to is just huge. I mean if it's safe and even

15 just a little bit efficacious for some people and we

16 could do it cheaply and broadly, you could imagine this

17 being added into the course of treatment in so many

18 current clinical cancer regimes, just to get a little

19 boost, just to get a little boost from the immune

20 system against that person's tumor in a way that seems

21 to be very safe. But we need to establish efficacy, or

www.transcriptionetc.com

190

1 this is all moot. But yeah.

2 DR. SANDUJA: I would like to add another

3 point of view to that. From a regulatory perspective,

4 if you look at that, we have gained and we have

5 gathered a lot of confidence in the platform approaches

6 that these are able to really very confidently, and

7 with a lot of robustness, can inform safety of these

8 products. And that's what has enabled a much faster

9 translation into clinical trials.

10 However, when it comes to approval or

11 licensure, that's a different question there. So we

12 are gathering confidence with respect to safety of

13 these platform approaches to further facilitate. And

14 that's being evaluated as we move forward with clinical

15 development of these products.

16 MS. WALKER: Hi. Karen Walker from Genentech.

17 Thanks for some really interesting conversations. I

18 have a couple of questions, again, going back to the

19 data and the bioinformatics. While I agree that safety

20 is a very important aspect -- and so is efficacy, also

21 so is supply and control -- once you have -- you're

www.transcriptionetc.com

191

1 treating lots of patients and the variability that you

2 described increases exponentially, how do you continue

3 to learn and identify what is important out of all of

4 the data that you're gathering versus what is just a

5 datapoint?

6 And I think that's a really important question

7 to ask and to answer. So I would be interested in your

8 thoughts.

9 DR. GRIFFITH: Yeah. I mean it's a hard

10 question. Do you think the increased variability from

11 doing the set scale on sort of a population scale, is

12 the implication there because there are so many players

13 in the space, and that's kind of a wild west of a

14 hundred different people doing it a hundred different

15 ways? Or do you just -- or do you mean more that we're

16 just servicing the tip of the iceberg in terms of how

17 patients respond and how different tumors behave?

18 MS. WALKER: I think it's more the latter.

19 DR. GRIFFITH: Okay.

20 MS. WALKER: But I also think it's what you

21 mentioned about the standard reference genome changing,

www.transcriptionetc.com

192

1 and what are disease-causing elements or the disease-

2 related elements versus just natural variability in our

3 own genetic sequences. This is the kind of thing that

4 I think we need to understand more.

5 DR. GRIFFITH: Yeah. I mean I think from a

6 personalized genomics aspect of it, personalizing it to

7 the reference genome of the individual, so that you're

8 comparing that against their tumor, that is a tractable

9 problem and one that has been tackled in other areas of

10 cancer genomics and NGS testing for cancer with tumor

11 genomes. In terms of just, yeah, how do we figure out

12 how to do it better in the face of all this variability

13 for different cancer types, I mean, I guess the high-

14 level answer, I would say, is to share the data openly

15 and let people have access to it and try to make the --

16 look inside the black box, that we really need to

17 understand what's going on in these pipelines, what the

18 process actually is, and what features are important.

19 All of the metrics, the readout, everything

20 needs to be exposed, so that we can -- because it's so

21 complicated it's going to be very difficult for one

www.transcriptionetc.com

193

1 group to understand. The pipeline is already so

2 complex that it's actually pretty much impossible for a

3 single person to really even understand the whole thing

4 from end to end. So it's really going to be a sort of

5 community team science or big team approach, even just

6 for the bioinformatics part of it. And that's just one

7 small piece of the complexity of this overall process.

8 MS. WALKER: Thank you.

9 DR. SAUNA: Could I follow with the question?

10 DR. GRIFFITH: Yeah.

11 DR. SAUNA: So to follow up on this question,

12 would it help if -- so you're already at some level of

13 precision by looking at an individual. As you get to a

14 deeper level of precision, looking at particular cell

15 types or single -- if you do single-cell sequencing,

16 and particular subsets of that tumor, which would

17 probably be more susceptible to the antigen and making

18 a tumor antigen targeting -- say a metastatic cell, for

19 example, rather than the tumor cells in general. Would

20 that level of precision help making it more effective?

21 DR. GRIFFITH: I think it's definitely

www.transcriptionetc.com

194

1 relevant to interpreting the response to the therapy

2 because the tumors are heterogenous.

3

4 And this is definitely a consideration of

5 ours, when we think about designing the vaccine, do we

6 specifically target only those antigens that are

7 thought to be in the trunk or a clone of the tumor that

8 will be in all of the tumor cells and not in a

9 subclone? Or is it, in some cases, okay to target

10 subclonal mutations? And then in terms of interpreting

11 the immune response, then the single-cell analysis of

12 the tumor microenvironment becomes very, very relevant

13 and useful potentially. But it's also very much a

14 developing area where there's so much to be learned.

15 MS. MCLELLAN: Hi. My name is Lorraine

16 McLellan. I'm a cancer patient and actual cancer

17 survivor. But I have something that's going to relapse

18 here in the next year or two. And I have done my

19 genome, and I am hopeful that I can do an neoantigen

20 vaccine.

21 But here we are in the headquarters for the

www.transcriptionetc.com

195

1 FDA. And what I would like to ask, because each of you

2 have done such an impressive job, is if I was able to

3 grant you one wish each to give a message to the FDA

4 today about what you would like them to do near-term,

5 and near-term, let's say 12, 18 months, to advance your

6 work individually, what would that be?

7 Would that be a question that I could each of

8 you to answer, so that we have a real takeaway and some

9 action items? Because we do have a bit of a need for

10 speed. I can appreciate that it's going to take a

11 couple years to get to gold standard. But what does

12 the FDA need to do near-term for each of you to advance

13 your work? Thank you.

14 DR. GRIFFITH: Do you want me to start? So in

15 terms of my individual work, I guess I would rather

16 answer more for the overall translation of this work to

17 patients. I think there is a challenge. And I've

18 heard this from quite a few representatives of -- in

19 research and in industry, that it's difficult to think

20 about developing a commercial version of a new antigen

21 vaccine without any clarity around whether the process

www.transcriptionetc.com

196

1 can be approved as safe, something that we're

2 comfortable with.

3 So I think that this meeting is very timely.

4 And I assume it is accompanied by an interest on the

5 part of the FDA, although I have no idea, to gather

6 information about how one might think of ultimately

7 giving some kind of regulatory oversight to these -- to

8 the process that's come up so many times. So yeah, I

9 would encourage the sort of continued serious thought

10 of that idea.

11 Because right now, I think it's a little bit

12 of an impediment for someone who's thinking about

13 trying to do this as a company that there isn't really

14 an obvious -- you know, you're not going to be able to

15 patent a drug. So -- and there's sort of risk averse

16 problem, where if they're -- if you don't have any sort

17 of stamp of approval that the process has been

18 evaluated in some kind of formal way as being safe,

19 then it makes it seem riskier to pursue it.

20 DR. SEYMOUR: I’ll go second. So I think it's

21 a really good question and one of the things I've sort

www.transcriptionetc.com

197

1 of put a hat on in trying to determine those different

2 aspects. So there is getting these therapies to

3 patients in a timely matter. And I think it sounds

4 like that is of critical need because a lot of these

5 diseases do progress very rapidly. So you don't have a

6 lot of time, particularly for the individualized

7 therapies.

8 And I think some of the discussion topics that

9 the FDA is having right now, I think are a path towards

10 that. And so I look at what does it take to get

11 something to a patient so that you can test it rapidly.

12 I think a lot of the pathways are already in place for

13 that in getting it to go into early-stage testing very,

14 very rapidly, whether it's individualized therapies or

15 not.

16 The second aspect is moving towards the

17 commercialization and licensure. And I kind of divide

18 those two separately when really thinking about

19 individualized therapies versus, say, bringing

20 therapies for a much larger population. I think

21 they're two separate questions. And so I think my wish

www.transcriptionetc.com

198

1 to the FDA is to continue discussions like this, work

2 with both industry as well as academic partners to come

3 forward with creative ways of bringing and testing

4 these products safely in patients and always keeping

5 in mind -- and I think everyone does on that -- that

6 benefit/risk and the severity of the indication that

7 we're trying to move forward with.

8 DR. JOUNG: Yeah. I don't have any specific

9 request for the FDA in terms of the work that we do. I

10 think for the community, as a whole though for gene

11 editing, I would encourage the community to have maybe

12 broader and wider-ranging discussions about how to

13 better standardize how to benchmark and how to develop

14 consistency around some of the safety and in particular

15 off-target testing because I think it's become very

16 fragmented. And so some of the issues that I raised in

17 my talk I think are things that we as a community need

18 to address.

19 And if we can do that, I think it will make it

20 easier for companies, academics, to be able to know

21 what it is that they need to do. Also, that it is an

www.transcriptionetc.com

199

1 evolving and fast-moving field, so the standards will

2 change over time. So to the extent that we can build a

3 mechanism by which there is -- I don't know exactly

4 what the form will be or what the body would be that

5 would do this -- but some kind of ability to respond to

6 changes as they occur, to be able to keep those

7 standards current and up to date. I think that would

8 be helpful for the field.

9 DR. SANDUJA: And to conclude that discussion,

10 I would like to -- from the FDA side -- would like to

11 reiterate that FDA acknowledges the challenges that are

12 associated with development of these individualized

13 drug products. And as we have seen, during all our

14 presentations and also during the discussion, there are

15 pathways and there are opportunities to discuss these

16 challenges and come to an agreement how they can be

17 resolved. Of course, we already agree that the

18 standard paradigm of drug development may not apply to

19 development of these individualized therapies. And as

20 the science behind these products is continuously

21 immerging and evolving, the Agency itself is open to

www.transcriptionetc.com

200

1 these discussions and further efforts, like the one we

2 are having today, to continue development of these

3 products. Thank you.

4 DR. RAYCHAUDHURI: I would like to thank all

5 the speakers and panelists and the audience

6 participants for what has been really an excellent

7 discussion this morning. And I'd like to thank Dr.

8 Sanduja for moderating this session. We are now at the

9 lunch break. And we have a very exciting afternoon

10 session ahead of us. So I ask that you please return

11 back to this room at 1:15, and we will proceed with the

12 afternoon sessions. Thank you.

13 [BREAK]

14 SESSION 3: CLINICAL

15 DR. RAYCHAUDHURI:

16 So I hope everybody had a nice break. So in

17 the morning sessions, we heard about challenges and

18 opportunities related to manufacturing of gene

19 therapies and phage therapies and tools for safety

20 testing and development of individualized therapeutics

21 products. This afternoon we're going to focus on

www.transcriptionetc.com

201

1 clinical development. The challenges and opportunities

2 to leverage what is known to facilitate development of

3 related products. And in session four, we will come

4 full circle on the focus and the reason for this

5 workshop, which is to discuss how we collectively can

6 increase access for patients to these critical products

7 in a timely and sustainable way.

8 So we have a very exciting agenda set for this

9 afternoon. I would like to introduce Dr. Rebecca

10 Reindel. Dr. Reindel is a medical officer in the

11 Division of Vaccines and Related Products Applications

12 in the Office of Vaccines Research and Review at CBER.

13 Dr. Reindel will be the moderator for session three,

14 which is on challenges and opportunities for clinical

15 development of gene therapy and phage therapy products.

16 Dr. Reindel.

17

18 SESSION 3 MODERATOR INTRODUCTION: DR. REBECCA REINDEL

19

20 DR. REINDEL: Thank you. I'm really excited

21 to be part of this third session. We have two

www.transcriptionetc.com

202

1 presenters. Dr. Schooley and Dr. Kohn will be

2 presenting, and then we'll have a panel session to

3 follow similar to the other sessions.

4 So by now you've seen several versions of this

5 slide. And I wanted to bring it back around to

6 clinical development and talk about the clinical

7 development of individualized therapeutics within this

8 paradigm. And as you can see, this figure describes

9 clinical development all the way through discovery and

10 pre-clinical, which we've covered quite a bit this

11 morning, and into the clinical phase of things where I

12 think this afternoon's sessions will sort of start to

13 take over.

14 Within this paradigm, typically you see

15 smaller Phase 1 studies that are designed to assess

16 safety and dose selection and then move up through

17 larger populations into Phase 2 and Phase 3 studies,

18 which study safety in an ongoing manner, and also start

19 to assess for effectiveness. However, individualized

20 therapeutics may not lend themselves as well to this

21 classic paradigm. And therefore, it's really important

www.transcriptionetc.com

203

1 that we look for opportunities for flexibility within

2 this paradigm with regard to clinical development.

3 In this session, we hope to identify both the

4 challenges that we face and anticipate in this space,

5 as well as any opportunities we can identify for

6 ongoing clinical development of individualized

7 therapeutics. So I like to think that every challenge

8 presents us with an equal opportunity to meet those

9 challenges and rise above them. So this is no -- by no

10 means an exhaustive list of all the challenges we face

11 in this space but some key highlights that we hope to

12 get to today as well as others include the following:

13 clinical investigations in the context of potential

14 manufacturing challenges, many of which were discussed

15 this morning.

16 In these studies, we need to pay a lot of

17 attention to study design when the product may be

18 different for every recipient or may need to be

19 tailored to a specific recipient or subject in real

20 time, such as to accommodate individualized treatment

21 of a patient with an infection that requires a specific

www.transcriptionetc.com

204

1 phage for that infection and then the impact of these

2 differences on the study procedures in the trial and

3 the interpretation of data that arises from these

4 trials. How do we interpret data that comes from

5 individual patients or subjects within the context of a

6 clinical trial that may include only maybe one or

7 several subjects? And also, how are we to interpret

8 novel endpoints? For example, micrologic- --

9 microbiologic endpoints for bacteriophage treatment.

10 This may pose some challenges in interpreting endpoints

11 in the absence of precedent.

12 Again, so what are the opportunities? And I

13 think a lot of the discussions today will focus on what

14 some of these opportunities are. So how do we create

15 infrastructure around a development program for an

16 individualized therapy for which there may be no

17 similar products developed or licensed in the past? An

18 example of this is, again, individualized bacteriophage

19 therapy where there's no existing model or structure

20 for clinical development or a specific guidance for

21 this type of product. And as experts in the field

www.transcriptionetc.com

205

1 emerge and collaborate, we can build systems that

2 facilitate the development of multiple research

3 programs. And we hope to address that today.

4 The roadmap of this infrastructure will

5 necessarily include novel clinical development programs

6 that may include innovative clinical trial designs and

7 statistical approaches to small population-based

8 clinical studies. Ideally, these early approaches to

9 building infrastructure can create a foundational basis

10 for future development. If we're able to leverage both

11 prior and collective experiences to guide us forward,

12 we can expedite and optimize the development of these

13 products that can meet specific individual needs.

14 So in order to maximize these opportunities

15 that we identify to meet the challenges that we see, we

16 will require flexibility. And where are there

17 opportunities for flexibility? Today, we hope to

18 discuss some innovative approaches to the clinical

19 portion of our product development program, including

20 areas where regulatory flexibility may be applicable.

21 Some examples of this include, as I mentioned, novel

www.transcriptionetc.com

206

1 clinical endpoints and statistical approaches that

2 allow for the enrollment of small populations of

3 subjects or subjects that may receive different

4 products within the same clinical study.

5 So some of the major areas that we hope to

6 cover today in our presentations and our panel

7 discussion include: the approach to designing and

8 interpreting efficacy assessments in studies that may

9 occur for a single individual or in a small group of

10 subjects. Again, the example of this is the

11 development of infrastructure around approaches to

12 demonstrate clinical benefits with phage therapy.

13 Along similar lines, accumulating safety data across a

14 range of disorders for treatments with genetically

15 modified hematopoietic stem cells can be informative.

16 Our presenters will also be addressing the

17 need for study designs that facilitate the

18 interpretation of clinical data that may present unique

19 challenges, such as the use of bacteriophages

20 adjunctive therapy in the context of complex and

21 varying antibiotic regimens across a variety of

www.transcriptionetc.com

207

1 anatomic locations and etiologic agents. Of course,

2 another important consideration, and many people have

3 already addressed this in the morning session, are the

4 timelines for development of much needed individualized

5 therapies. Today, we'll discuss the impact of these

6 timelines on end-to-end development and approaches to

7 optimizing this timeline in the context of advanced

8 therapies.

9 As we move from our presentations into our

10 panel discussion, we hope to touch on the some of the

11 following points, and we're looking forward to a

12 thoughtful and insightful discussion with our patient -

13 - with our panelists, speakers, and audience members.

14 Some of these include the approaches that may

15 facilitate the assessment of efficacy in

16 individualized therapeutics, especially in the context

17 of small groups of subjects. Are there ways to

18 leverage accumulated safety data to enhance assessments

19 of safety? How do we approach the interpretation of

20 safety data in clinical development programs that may

21 not progress according to the usual paradigm? And what

www.transcriptionetc.com

208

1 are some statistical and study design approaches that

2 we can consider as we exercise flexibility?

3 So I'd like to introduce our first speaker.

4 This is Dr. Schooley, who currently serves as Professor

5 of Medicine and Senior Director of International

6 Initiative at UCSD, one of my alma maters. He

7 completed medical school and an internal medicine

8 residency at Johns Hopkins and an ID fellowship at NIH

9 and Mass General Hospital. He was at Harvard in 1981

10 with early research efforts directed at the

11 pathogenesis and therapy of herpes group and retroviral

12 infections. He was head of the Division of Infectious

13 Diseases at University of Colorado in 1990, another one

14 of my alma maters. And he led the NIH AIDS Clinical

15 Trials Group from 1995 to 2002. And he's currently, as

16 I said, now at UCSD, and he's serving as the head of

17 UCSD's Infectious Disease Division. And his recent

18 interests have focused on the use of bacteriophage to

19 treat infections. And we are excited to hear his talk

20 today.

21

www.transcriptionetc.com

209

1 OPPORTUNITIES AND CHALLENGES IN THE CLINICAL

2 DEVELOPMENT OF BACTERIOPHAGE THERAPEUTICS -

3 DR. ROBERT T. SCHOOLEY

4

5 DR. SCHOOLEY: Thanks very much. It's a

6 pleasure to be -- to follow someone who's had such an

7 illustrious pathway in terms of institutions she's been

8 in and to try to talk to you today a bit about a

9 juncture in phage therapeutics. I'm going to try to

10 bridge some of the elegant discussion you heard this

11 morning from Jason Gill about some of the aspects of

12 phage production, phage biology, say a little bit about

13 the -- where phage therapeutics are today and then to

14 go on to reach forward to the final discussion this

15 afternoon where we'll be talking about access to

16 emerging therapeutics with a few comments about how

17 those might be able to fit into some of the clinical

18 trial development approaches as well.

19 So, the -- today, I'm going to try to briefly

20 talk about some of the limitations of antibiot- --

21 anti-microbial therapy. Everyone here is quite aware

www.transcriptionetc.com

210

1 of most of that, say a little bit about how phage

2 therapeutics might fit into some of these gaps. I'll

3 say a small amount about what we know today, go on to

4 talk about some of the key gaps, and then talk about

5 moving from where we are today to orderly clinical

6 development.

7 Obviously, these days as you read the

8 increasing number of reports of multidrug resistant

9 bacterial infections, it's clear that the microbial

10 evolution on a global platform is outpacing our ability

11 to keep up with them with small molecule traditional

12 antibiotics. So one of the obvious places for phage

13 therapeutics is to deal with this because they've been

14 innovating in terms of any microbial activity for about

15 300 million years, and we only started about 80 years

16 ago. The -- we also know that phages do some things

17 that antibiotics don't do, for example, interfering

18 with biofilms. We know that phages can go to some

19 places that are difficult to penetrate with antibiotics

20 and places where antibiotics don't work as well. So

21 they're -- in addition to just spectrum, there's some

www.transcriptionetc.com

211

1 other aspects of phage therapeutics that are attractive

2 in thinking about how we might take advantage of these

3 activities in individualized applications.

4 So what are the opportunities here? There's a

5 virtually unlimited number of and diversity of phages

6 in nature. And we have tools now to genetically modify

7 those phages. Whether we will need to do that and what

8 we can do to make them better than they already are, I

9 think, remains an area of intense interest and one that

10 will have to be validated in the clinic in a series of

11 iterative clinical studies.

12 We know that there's some opportunities with

13 phages that are quite attractive, particularly the

14 pharmacodynamics. With antibiotics, we give a dose of

15 antibiotic, and we're already worried about whether the

16 kidneys or the liver or both are leaching the

17 antibiotic away from the site of infection before the

18 next dose. With phages, in theory at least, once the

19 site of infection is seeded, self-replication can

20 continue to provide a phage to deal with the

21 antimicrobial challenge.

www.transcriptionetc.com

212

1 We also know that phages can disrupt biofilms.

2 There have been an increasing number of animal studies

3 and clinical anecdotes in which this seems to be

4 playing a role. Phages have a theoretical advantage of

5 being less disruptive to the microbiome and

6 contributing to further antibiotic resistance when

7 they're used in -- as targeted therapeutics. And

8 finally, there are some reports of how phages can re-

9 sensitize organisms to antibiotics through both

10 mechanism-based approaches and more generally.

11 Now, where would you think about using phages?

12 Obviously, for patients that have organisms that are

13 not susceptible or to antibiotics that patients can

14 tolerate, places where we're not -- where antibiotic

15 delivery or activity is limited by the anatomy, and in

16 situations in which having something to deal with

17 biofilms might be particularly attractive. And these

18 are areas that are already under investigation and also

19 targets of individual experiences in the clinic.

20 So what do we know today? Well, we know this

21 morning that -- from Dr. Gill's comments, we can make

www.transcriptionetc.com

213

1 relatively large batches of phages quite safely and

2 quite uniformly. And we can administer them to the

3 patients with very little evidence of toxicity if the

4 phages are produced in ways that are cognizant of

5 contaminants and deal with issues related to genes you

6 might not want to have in the phage and so forth. So

7 the technology is there to produce phages in a very

8 homogenous way. There are a fair number of nuances

9 about phage stability in different conditions and host

10 strains and so forth that are quite different from the

11 antibiotic situation. But they're all solvable in this

12 context of individualized therapy.

13 We know that there have been a lot of -- a

14 large number of anecdotal cases in which phages seem to

15 have shown benefit for individual patients. But these

16 are all anecdotes. And we need to move from anecdotes

17 to more organized data to be able to understand how to

18 actually use phages in clinical practice. And we know

19 that, as with antibiotics, resistance develops quickly.

20 And we need to understand how to circumvent this with

21 phage cocktails and other approaches that are also

www.transcriptionetc.com

214

1 under development.

2 So what are the gaps? Well, one of the major

3 gaps is how do we know which phage or phage combination

4 is best to use in a given patient? If one remembers

5 back to the early antibiotic days, early antimicrobial

6 testing involved agar plates, broth dilutions. And we

7 spent a long time, 20 or 30 years optimizing predictive

8 mod- -- approaches for clinical efficacy, and we’re

9 still not there. Phages are the same story.

10 We have very little knowledge about what the

11 optimal dosing levels, routes of administration, and

12 duration of therapy should be with phage therapeutics,

13 the classical Phase 2 sorts of data that one would want

14 to have of an antibiotic before proceeding to clinical

15 endpoint trials. We don't know enough about the

16 antimicrobial activity of phages in vivo in humans. We

17 know that they can select for resistance, which is a

18 very good measure of demonstrating they have any

19 bacterial activity. But they -- how the antimicrobial

20 activity compares, for example, to antibiotics head-to-

21 head is something that is still up in the air.

www.transcriptionetc.com

215

1 We know that resistance develops quickly, but

2 we don't understand the determinants and the kinetics

3 of resistance and how that may be affected by things

4 like phage host range and other aspects of phage

5 biology. And we need to understand how to mitigate

6 this. And finally, we need to move on to think about

7 how to demonstrate phage efficacy in specific clinical

8 situations.

9 Now, where is phage therapy these days? Well,

10 there's been a lot of off-line use for over 100 years

11 in many parts of the world. And I think the take home

12 message for most of this is that, by and large when

13 phages are given orally or topically, there's very

14 little evidence of toxicity. But it's been very

15 difficult to assess objectively whether there's any

16 evidence of efficacy because many of the endpoints are

17 chosen post-hoc.

18 Many of the patient populations tested were

19 not homogenous, and very few of the phage preparations

20 used were well-characterized. We know that there are

21 increasing number of individual uses under eINDs. And

www.transcriptionetc.com

216

1 again, we're beginning to see that some of these are

2 becoming more organized and more standardized, in some

3 ways bridging the way to early clinical trials from the

4 standpoint of a little more homo- -- homogeneity.

5 So how might this eIND experience bridge

6 towards IND experiences? Well, one approach is that as

7 people who are confronting patients along with the --

8 often with similar clinical indications confer with

9 each other, we often will converge on the same

10 approaches and based on what other people have done so

11 that each patient isn't a brand new patient. I'll talk

12 to you a little bit about how the iPATH center at UCSD,

13 which has begun to both do clinical trials and assist

14 with clinical cases, has been trying to do that.

15 We also know that with -- standardized

16 clinical approaches do emerge when people do the same

17 thing over and over again. There was a very nice case

18 series in the Nature Microbiology that appeared in

19 paper this month and online last month about a dozen or

20 so patients with staph sepsis from Australia. And the

21 M. abscessus experience with Graham Hatfull, since

www.transcriptionetc.com

217

1 Graham is about the only one who has M. abscessus,

2 we've been collaborating with him. And most of the

3 therapeutic interventions there have been pretty

4 standardized because we really don't have many data

5 points to start variation.

6 Now, how have we tried to do some of this at

7 iPATH in terms of trying to, while the clinical trials

8 are up and going, make phages available to people who

9 need them? We have been engaged in this -- initially

10 stumbling into it about four years ago with a faculty

11 member at UCSD. And then one of our astute clinicians

12 began to identify other patients who needed phage

13 therapy. And since that time, another eight or nine

14 patients have been treated at UCSD. And then, we began

15 to get calls from other places about how to approach

16 phage therapeutics. And the Chancellor of UCSD put

17 together some seed money for us to put an

18 infrastructure together to be able to respond to some

19 of these requests as they came in.

20 The approach we currently take is, when we get

21 a request from a patient or family or treating

www.transcriptionetc.com

218

1 physician, is to try to get back to the treating

2 physician because we are not trying to be the patient's

3 doctor. And many times in those settings, things get

4 lost in translation, and we don't want to be giving

5 medical advice to people who are not in front of us.

6 After we talk to the doctor, often the doctor

7 will tell us what this patient needs is a dose of

8 ampicillin, or what this patient needs is nothing. And

9 we move on, or sometimes the physician will say, “You

10 know, I don't really know what else to do but would

11 phage therapy be reasonable?” If it seems reasonable

12 we'll talk through some of the options. And if the

13 physician wants to proceed, then we proceed to try to

14 help them do that, with the first step being trying to

15 help them find a phage product that can be used.

16 There aren't many sources for those these days

17 in terms of phage production and discovery operations

18 that provide phages of sufficient quality that we would

19 feel comfortable giving to patients in any kind of an

20 organized way. We have kind of -- there are a group of

21 collaborating institutions and investigators who have

www.transcriptionetc.com

219

1 expertise with specific organisms. People at Texas A&M

2 are particularly good at Burkholderia. Baylor works on

3 E. coli and Klebsiella. Yale has been very interested

4 in Pseudomonas and so forth.

5 And so what we'll often do is say to a

6 physician, “We don't have any solution for you, but you

7 might want to talk to Jason Gill or to someone at one

8 of the other places.” They will then send an isolate

9 to that location, and they will then -- at this

10 location, if they have agreed to try to screen, will

11 either come up with a phage or not. So one of the

12 bottlenecks is whether or not the receiving laboratory

13 can come up with a phage that -- or phage, preferably a

14 cluster of phages that might be active against the

15 patient's isolate.

16 If they find a suitable phage, there are a

17 couple of -- the next bottleneck is moving from an

18 academic laboratory discovery operation to being able

19 to have a product that be given to a patient

20 perennially. And many academic labs haven't done that

21 before. They have been -- they have very high-quality

www.transcriptionetc.com

220

1 operations. But things -- thinking about pharmacologic

2 stability, how to dispense the product over a period of

3 days or weeks from an investigational pharmacy, dealing

4 with things like bacterial contamination with a USP 71

5 testing and so forth, these are not things that many

6 academic labs think about. And one of the things we've

7 tried to do is help them at least become aware of

8 those.

9 And increasingly, these labs have -- are now

10 incorporating that so that when they talk to the FDA in

11 conjunction with the referring physician -- because

12 that's where the eIND has to come from -- many of these

13 things have already been taken care of before the FDA

14 reviewer has to say, “So what have you done about

15 sterility?” If they ask us about dosing we say, “We

16 don't know the answer to that, but this is what we've

17 done in the past. And these are things you might

18 consider.”

19 And what often happens is people will use

20 doses that are similar, routes that are similar to what

21 we've used before. And while clinical trials are

www.transcriptionetc.com

221

1 proceeding and being developed, we hope that some of

2 this homogeneity will help, at least within the agency,

3 begin to look at cases in more of a -- in the aggregate

4 than as each case being a different approach with

5 different doses given by different routes for different

6 durations. And the phage therapy that is given to the

7 patient is obviously given -- provided by the referring

8 -- by the laboratory and given under eIND.

9 So another thing that is happening under this

10 is, as time has gone on and more of these experiences

11 have evolved, there are certain very frequent flyers

12 that come up, including treatment of non-tuberculous

13 mycobacterial infections. Graham Hatful has been

14 collaborating with us for about two-and-a-half years

15 now and has had about 90 requests for treatment, mainly

16 of M. abscessus, mainly in trans- -- renal transplant,

17 liver transplant patients, immunocompromised patients.

18 And the bottleneck there has been mainly finding

19 isolates that are -- phage that are active against any

20 given isolate.

21 Having said that, because the patients are

www.transcriptionetc.com

222

1 similar, the requests are similar, we've developed a

2 shell protocol that we're currently trying to refine

3 and work with people at the NIAID to begin to think

4 about how this might move on into a real clinical trial

5 but, in the meantime, suggesting approach like this

6 when patient -- when physicians want to take this to

7 the FDA as eINDs before this emerges. Now, where do

8 you go from there? Well, it's not rocket science about

9 how to develop antibiotics, and these are antibiotics.

10 I think one of the things that has happened

11 with phage therapy is that it has been treated as if

12 it's something else. These are antibiotics; they just

13 happen to be living. And we've had paradigms to

14 develop antibiotics for 80 years. And if we don't use

15 those paradigms, taking into account some of the

16 biological variabilities, differences with phages, we

17 won't know how to use them in clinical practice.

18 And we won't know how to benchmark them with

19 antibiotics when choices are made. And we won't know

20 how to use them together because phages will not be

21 used instead of antibiotics except in some

www.transcriptionetc.com

223

1 circumstances. At least initially, they will be used

2 with antibiotics. We need to use them in a context

3 that we understand. And we developed -- we,

4 collectively as a community, have developed approaches

5 to evaluate antimicrobials.

6 Now, there are some nuances to phage

7 therapeutics that are different, and you have to think

8 about in terms of the typical Phase 1, Phase 2, Phase 3

9 design and issues related to CMC. We've -- some of

10 which were talked about this morning by Jason Gill. So

11 the clinical trials are -- have begun and one of the

12 challenges has been trying to think about how they can

13 be done in an orderly way.

14 You've seen most recently what amounted to a

15 Phase 3 trial done in Europe on burn patients before

16 Phase 2 data had been generated in terms of

17 understanding dosing, understanding stability on the

18 way to the patient, understanding phage interactions,

19 and even understanding the microbiology of what was

20 being treated. So we've had a lot of enthusiastic

21 efforts to get ahead of the curve without going through

www.transcriptionetc.com

224

1 the steps between here and there to develop the kind of

2 data to have studies be done in a more organized way

3 that you would do if you had an antibiotic.

4 Now, Phase 1 trials, if you start going

5 through the typical approach with phage therapeutics, I

6 would argue are almost useless at this point. We know

7 phages are basically safe if they are prepared well.

8 It doesn’t make a lot of sense to try to understand

9 pharmacokinetics and pharmacodynamics of phages in

10 patients that don't have an organism the phage will

11 grow in. So the traditional, “But have you given that

12 to a human yet,” whenever a new phage comes along is a

13 waste of time to talk about and really, I would argue,

14 don't help much anymore.

15 Moving to Phase 2, the major caveat I would

16 make about this is in -- with antibiotics we would

17 demand Phase 2 data before going to Phase 3. And if

18 any generalization can be made about drug development,

19 going to Phase 3 before you understand Phase 2 has been

20 a graveyard for drugs of all classes. And I think

21 phage therapeutics is one that needs to be careful not

www.transcriptionetc.com

225

1 to fall into that trap.

2 So one of the Phase 2 trials that is being

3 developed is one that is being supported by NIAID and

4 being carried out by the Antibiotic Resistance

5 Leadership Group. It's a very simple study trying to

6 understand the activity of phages in humans outside the

7 context of when antibiotics are given in conjunction.

8 This is a study that would be done in patients with

9 cystic fibrosis, who are clinically stable, and don't

10 need antibiotics at the time but are shedding

11 Pseudomonas aeruginosa chronically.

12 The inclusion criteria would be mainly -- we

13 would be looking at people who repetitively shed

14 pseudomonas in their sputum. There are quite a few

15 people like this who are clinically stable. The goal

16 here would be to do a standard single ascending dose

17 study to understand when phages are given by an

18 intravenous or by an aerosolized route, how long they

19 reside in the lung, what they're antimicrobial activity

20 is, what the evolution of the phage and the organism is

21 under treatment when used together, and to use that to

www.transcriptionetc.com

226

1 build a database to move on to do multi-dose studies

2 and studies along with antibiotics in Phase 3 trials.

3 It's a very similar -- very simple design of multiple

4 cohorts with a placebo in each cohort and, at the end

5 of the day, expanding the cohort that looks optimal to

6 get more precision around the measurement.

7 Now, Phase 3 trials are moving along. I've

8 already made the point that we need to, I think, be

9 careful about launching Phase 3 trials unless we

10 understand what we're doing because the worst thing for

11 this field would be several failed Phase 3 trials that

12 are done in a way in which the data were

13 uninterpretable. The -- it didn't work. See, they've

14 been trying for 100 years, here's another failure is

15 probably the biggest, I think, short term danger to

16 this field because it may or may not have promise. But

17 it would be a shame to have it put aside without

18 understanding the science under the hood.

19 So in terms of Phase 3 trials, I've already

20 made this point. These studies should be done as if

21 these are antibiotics. That's what they are. And we

www.transcriptionetc.com

227

1 need to have microbial endpoints. We need to put them

2 together with clinical endpoints. We need to think

3 about them in the kinds of trial designs that one would

4 use for treatment of multi-drug resistant organisms, if

5 that's what you're after.

6 We can think about other clinical

7 applications: implanted prosthetic devices in which you

8 might be trying to show, in a placebo-controlled way,

9 that you can salvage devices that would otherwise have

10 to be removed. There are many clinical trial designs,

11 but they should be thought about in ways that give

12 crisp endpoints and that are -- give you information

13 that will help you clinically.

14 In these Phase 3 trials, the unique aspects

15 that are different from antibiotics are that they --

16 the organism that the antibiotic in question actually

17 replicates after you give it at the site of infection.

18 And I think mathematical modeling will be very

19 important was we move ahead to understand relationships

20 between population sizes and clinical scenarios. We're

21 going to be in a situation where we will have to think

www.transcriptionetc.com

228

1 about aggregating patients with specific clinical

2 conditions, like E. coli UTIs, in clinical trials in

3 which each patient will be treated with a different

4 agent. And by using endpoints that are well-defined

5 and agents that are well-defined in terms of how they

6 are made, we should be able to develop systematic

7 information that will inform clinical use.

8 Finally, people have talked about phage

9 therapeutics as being high risk, high gain. I would

10 argue that's not really the case. They really are

11 potentially high gain, but are they really high risk?

12 Phage have been around for 300 million years, and

13 they're still here killing antibiotics -- or killing

14 antibiotics -- not yet. They're still here killing

15 bacteria. And what we've had over the last 15 or 20

16 years is a lot of advancement in terms of how to

17 prepare phage, how to work with them in laboratories,

18 how to purify them, and get them to the bedside.

19 And what we need to do now is to develop

20 rigorous clinical trials to figure out how best to use

21 them and to have us understand that the reason we do

www.transcriptionetc.com

229

1 clinical trials isn't to get drugs approved; it's to

2 learn how to use them in clinical practice. And so of

3 course the optimal clinical trial is when you can do

4 both at the same time, and I think phage therapeutics

5 is ripe for that at this point. Thank you very much.

6 DR. REINDEL: So our next presentation is

7 going to be conducted remotely due to travel issues.

8 Dr. Kohn is a professor in the Departments of

9 Microbiology, Immunology, and Molecular Genetics as

10 well as Pediatric Hematology/Oncology at UCLA. He's

11 board certified with more than 30 years of experience

12 in treating children in the clinical

13 transplantation space.

14 His principle area of research is the

15 development and application of methods for gene therapy

16 of blood cell diseases using autologous hematopoietic

17 stem cells. His lab has investigated methods for

18 optimal gene delivery and expression and gene editing

19 with human hematopoietic stem cells performed in

20 clinical trials of gene therapy for genetic diseases

21 and pediatric HIV and AIDS. He's won many awards and

www.transcriptionetc.com

230

1 been appointed to many prestigious positions, and we're

2 excited to hear his talk today.

3

4 CHALLENGES TO DEVELOPING INDIVIDUALIZED STEM CELL GENE

5 THERAPIES - DR. DONALD B. KOHN

6

7 DR. KOHN: Well, thank you. Can you hear me?

8 Okay. Thank you. Talking to a phone, it's hard to

9 know. Sorry, my travel issue isn't a virus. It was

10 multiple mechanical problems on United Airlines that

11 couldn't get me there.

12 So I'm going to talk about work that we've

13 done over the last couple of decades actually to

14 develop gene therapy for a rare disease, ADA SCIDs that

15 I'll talk about, kind of, to show that, maybe, if

16 anything, this is the old school traditional route and,

17 at the end, talk about ways that we can possibly bring

18 these kinds of therapies about more quickly forward.

19 So the next slide is my disclosure. And it's relevant.

20 I'm on the board for a company called Orchard

21 Therapeutics, and my university has licensed IP to

www.transcriptionetc.com

231

1 them. And I'll talk about that today. That's the ADA

2 SCID gene therapy. Next slide.

3 So what I'm going to talk about is just give a

4 little background on sort of this area of therapeutics

5 targeting hematopoietic stem cells with gene therapy to

6 treat blood cell diseases. Then I'll talk about it as

7 a case example gene therapy for ADA SCID. And then

8 I'll close with some not so deep insights on lessons

9 that are learned that might be useful for development

10 of individualized therapies. Next slide.

11 So this is required by our union to be shown

12 at all presentations. This is a hematopoietic tree,

13 making the point that it's the hematopoietic stem cell

14 that lives normally in our bone marrow that both self-

15 renews and gives rise to all the blood cells. Next

16 slide.

17 And so hematopoietic stem cell transplants can

18 cure a whole list of genetic diseases of blood cells,

19 and this is now over 40, 50 years of work. We can take

20 allogeneic stem cells from a well-matched donor, or

21 even a haplo-identical donor now, and transplant them

www.transcriptionetc.com

232

1 to essentially replace the patient's own stem cells,

2 burying the monogenic defect to treat classes of

3 primary immune deficiencies. And I'll talk about SCID,

4 hemoglobinopathies like sickle cell and thalassemia, a

5 whole list of lysosomal storage and metabolic and

6 , and congenital cytopenias.

7 So they -- it won't -- you can't treat

8 everything with hematopoietic stem cells, but there's

9 at least several dozen blood cell related diseases that

10 are macrophage, monocyte-related disorders that can be

11 treated by replacing the defective stem cells. Next

12 slide. And so, again, showing on this tree, so then

13 the technical task is that the gene correction event

14 needs to occur in the multi-potent long-term

15 hematopoietic stem cells. Everything after that is

16 sort of entrenched and amplifying effect cell, and the

17 effect would be short-lived if we put the gene into,

18 for example, a progenitor.

19 And so the two sort of major approaches that

20 have developed now are adding a gene using an

21 integrating virus, as I'll show you. Or the really

www.transcriptionetc.com

233

1 exciting emerging area that Keith touched on a little

2 bit in the therapeutic approach is to edit the gene in

3 the stem cell. And either of these if you make a

4 permanent change in the genome of the stem cell that

5 will then be propagated to all the blood cells that

6 follow. Next slide.

7 And so this is cartoon, sort of, the process.

8 And it starts with the patient. And their

9 hematopoietic stem cells are isolated, which is one of

10 the things that makes this therapy much easier than all

11 the other in vivo approaches that we take the cells out

12 of the body, then in the laboratory either add the gene

13 with a integrating vector and several of the types of

14 viruses that are listed there, or use the new -- all

15 the gene correction methods to site-specifically

16 correct the defect or knock out a gene or change a base

17 or whatever you want to do in the stem cells ex vivo.

18 And then, typically before the cells are given

19 back to the patient, the patient will receive some

20 types of chemotherapy, or in the future hopefully,

21 monoclonal antibodies to get rid of some of their own

www.transcriptionetc.com

234

1 hematopoietic stem cells to make space so that when you

2 give back this modest amount of cells that you've take

3 from the patient they can reconstitute a lot of their

4 blood cell production. Next slide. And so the field

5 has gone through sort of two major rounds of viral

6 vectors. And so the first generation of vectors shown

7 at the top were from -- typically from murine Moloney

8 virus, where the virus' long terminal repeats

9 were intact and had strong enhancer promotors that make

10 a lot of the transgene messenger RNA and transgene

11 protein then. But these were dangerous because they

12 had strong enhancers in their LTRs that can

13 transactivate an adjacent gene so that, when you add

14 these cells to 10 to the 8th cells from the patient's

15 bone marrow, they land relatively randomly. And if

16 they happen to land next to a proto-oncogene, the

17 enhancers could turn them on. And that in fact

18 occurred in some of the clinical trials in the 2000s.

19 So the field has largely turned to sort of the

20 types of vector shown at the bottom, these second-

21 generation self-inactivating, or SIN vectors, where the

www.transcriptionetc.com

235

1 enhancers are deleted from the long terminal repeats.

2 And then the gene can -- transgene can run off an

3 internal promoter that can be selected, either that

4 it's a reasonably strong promoter without a lot of

5 enhancer activity or, in fact, can be lineage specific

6 like beta globin to make beta globin for sickle- --

7 thalassemia or sickle cell. Next Slide.

8 And so using this approach -- and this lists a

9 number of the disorders now that have been treated, not

10 approved drugs yet but at least in preliminary Phase 1

11 and 2 trials, show evidence of clinical efficacy and

12 good safety. So many of the diseases that were on that

13 initial list have now been approached. And one of the

14 challenges here that, you know, is even more extreme in

15 individualized therapies is that each genotype requires

16 a separate vector carrying the gene and a developmental

17 project.

18 And so it sort of limits the development and

19 the number of disorders that are approached. But in

20 fact, the safety record from these vectors to the

21 present time has been quite good. There have been no

www.transcriptionetc.com

236

1 vector related serious adverse events that I'm aware

2 of. And one of the worries has always been, since

3 you're using a vector based on HIV, the potential for

4 elements of the vector and the packaging that come

5 together to make replication confident lentiviral

6 vectors that could spread. And to my knowledge again,

7 this has never been reported in either products or in

8 patients.

9 And using the lentiviral vectors, the

10 integration sites don't show preferential integration

11 near oncogenes. And there have not been any clinically

12 significant clonal expansions, again, that I'm aware

13 of. So they're looking relatively safe, although still

14 it's probably maybe 500 people or maybe 1,000 worldwide

15 that have received these kinds of vectors into

16 hematopoietic stem cells. So it's still relatively

17 early in the developing a safety base. Next slide.

18 And so then I want to talk about what's been

19 my favorite disease to treat for many years now, Severe

20 Combined Immune Deficiency or SCID. And SCID is the

21 most severe of the human primary immune deficiencies,

www.transcriptionetc.com

237

1 of which there's several hundred. SCID has absent T

2 and B cells, and NK function is variable depending on

3 the genetic type. And in fact, SCID can be caused by

4 more defects than any -- in any one of more than 20

5 genes. And in total, SCID is quite rare. About 1 in

6 58,000 is one of the best estimates from -- now that

7 there's newborn screening we're getting a much better

8 feel for the frequency. And SCID as a severe immune

9 deficiency has been uniformly fatal in infancy before

10 treatments were developed.

11 Typically, there'd be severe recurrent

12 infections, chronic diarrhea, failure to thrive leading

13 to death in infancy. And there was one famous child

14 who was maintained in a germ-free bubble for more than

15 a decade. And that's why it's sometimes called bubble

16 baby disease.

17 And we know that bone marrow transplant can be

18 curative. So we know that giving normal hematopoietic

19 stem cells can essentially replace the immune

20 deficiency. And in fact, if there is an HLA-matched

21 sibling donor, which occurs in about 20 percent of

www.transcriptionetc.com

238

1 patients, there's more than a 95 percent success rate.

2 And the small failures are typically patients who have

3 severe infections at the time of transplant. But for

4 the majority of patients that don't have a matched

5 sibling, the results have been less good. Although,

6 they continue to improve using either matched unrelated

7 donors or haplo-identical typically parental donors.

8 Next slide.

9 And so then, just the specific disorder that

10 we focused on, ADA SCID, it's the cause of about 10 to

11 15 percent of human SCID. So it's the second or third

12 most common gene that can cause SCID. And we estimate

13 there's about 10 children born a year in the U.S. and

14 Canada based on the referrals that we've had and the

15 population incidences. And ADA SCID patients have

16 profound pan-lymphopenia. So they have typically --

17 essentially no T, B, or NK cells at shortly after birth

18 from accumulating the toxic adenine metabolites that

19 ADA would normally be part of catabolizing.

20 So ADA SCID has been the focus because it was

21 the first genetic form of these more than 20 of human

www.transcriptionetc.com

239

1 SCID with a biochemical. And then the genes were

2 cloned sort of in the mid-'80s. And so it's the first

3 where things began because the gene was in hand. And

4 so as I referred to there, there are multiple

5 therapeutic options for patients including all the

6 allogeneic stem cell transplants from matched siblings,

7 matched unrelated, or haplo-identical donors.

8 There's also an FDA approved, and in fact two

9 serially, a purified one and now a recombinant, enzyme

10 replacement of polyethylene glycol modified ADA that

11 can be used to lower systemic ADA levels. And then

12 there's also emerging autologous stem cell transplant

13 gene therapy that I'll talk about. So the next slide

14 shows the lentiviral vector that we've worked with now

15 for the last seven, eight years.

16 So we've been doing trials sort of

17 successively over 20 years using the earlier type of

18 vectors that I showed you. And then about eight years

19 ago, talking to colleagues at the University College

20 London, Adrian Thrasher and Bobby Gaspar, we said we

21 should move to a lentiviral vector, and this one was

www.transcriptionetc.com

240

1 developed. So it has the human ADA cDNA running off

2 the elongation factor alpha core promoter, which is one

3 of those promoters I mentioned. It's a pretty strong

4 promoter but doesn't have much transactivating activity

5 of trans enhanced nearby genes.

6 And this turned out to be a very well-behaved

7 vector. So it's one of the SIN types of vector with an

8 elongation factor promoter. The cDNA is codon

9 optimized to get better expression. The WPRE elements

10 stabilizes the method so you get more bang for the

11 buck. You get more protein per transcript, and it's

12 pseudotype VSV-G. And it has a very high titer. This

13 is from our lab. We can get very high titers after we

14 concentrate it, so it goes into stem cells very

15 efficiently. Next slide.

16 And so we spent about two years once we had

17 this vector chosen doing the pre-clinical work that

18 comprised the IND pharmtox package. And so we looked

19 at efficacy in term of the transfer and expression of

20 the ADA gene in patient derived bone marrow cells, both

21 in vitro, and then put into immune deficient mice in

www.transcriptionetc.com

241

1 vivo. And compared to a retroviral vector that we were

2 using before, MND ADA, the EFS ADA lenti had higher

3 gene transfer and higher ADA production per vector copy

4 number. So we had good activity.

5 And then we did a series of safety studies.

6 We put this vector into bone marrow of either ADA

7 deficient mice or to human cells in immune deficient

8 mice. And in fact, either the retro or the lenti,

9 neither showed any leukemia or clonal expansion in

10 these models, maybe suggesting these models are not

11 very robust. Integration set analysis did show that

12 the retroviral vector was more often near the

13 transcriptional start sites in cancer related genes

14 that the lentiviral vectors.

15 That's kind of a very well-known recurrent

16 pattern of integration for these two classes of

17 vectors. And in fact, in an in vitro assay, the murine

18 gamma retroviral vector caused murine lineage negative

19 bone marrow cells to clonally expand using an assay

20 called in vitro mutagenesis assay and the lentiviral

21 vector didn't. And so in fact, we had used the gamma

www.transcriptionetc.com

242

1 retroviral vector that we're comparing here for several

2 trials beforehand and have had no adverse events from

3 the vector. But these pre-clinical studies showed the

4 lenti was at least as safe and possibly safer. And so

5 that's sort of those studies which we published

6 composed the toxicology and pharm package for the IND.

7 Next slide.

8 And so I just wanted to take -- you know,

9 what did it take to do? So this is the slow road to

10 lenti, which is a pun. And so this shows you in the

11 academics what it took us to get from, “Hey, we should

12 make a lenti” to an open trial. And so the gree- --

13 highlighted in green are the funding applications where

14 we got funding in various stages to do the -- pre-

15 clinical work was done on a program project grant we

16 had from heart, lung, and blood. Heart, lung, and

17 blood also had a gene therapy resource program that

18 paid for the GMP comparable vector for the pharm-tox

19 study. And then the NIAID, we received an U01 award

20 for the clinical trial, for the Phase 1 trial.

21 And we went through a regulatory gauntlet of

www.transcriptionetc.com

243

1 the RAC, the FDA pre-IND. This study was initially

2 opened up for both UCLA and the NIH. So we had IRBs

3 and IBCs at both places. And then, so we submitted the

4 IND at the end of 2012, so about four years from sort

5 of proof of concept to an IND. And I think probably

6 that can be done more quickly. We were sort of

7 learning as we were going. Next slide.

8 And so in fact, we opened up this Phase 1

9 trial in the U.S., and a parallel trial opened up in

10 London where we treated patients with this vector in

11 low dose relatively Busulfan conditioning. And we'd

12 roll patients who had ADA SCID without a matched

13 sibling donor. They had adequate organ function and

14 could not have an ongoing active infection.

15 And our primary endpoint was safety, survival,

16 event free survival, event being, sort of, failure and

17 needing to have a rescue transplant, and scored adverse

18 events. And then secondary endpoints were more for

19 efficacy, measuring the production of gene marked blood

20 cells from the stem cells we treated, looking at immune

21 reconstitution, and clinical endpoints of infections,

www.transcriptionetc.com

244

1 and hospitalization. So the trial opened up in May

2 2013 under an IND. Next slide.

3 And we actually went through and treated

4 patients relatively quickly. The initial plan was to

5 treat 10 patients, but we kept getting referrals. So

6 we wound up treating 20 patients. So over the course

7 of about five years, we went from the first patient

8 into the last patient visit treating 20 of the ADA SCID

9 patients. Next slide.

10 And these are some early interim data. These

11 aren't the official data, but these are data when

12 about, I think, 15 of the patients had been -- were out

13 at least a year, looking at various outcome parameters.

14 And so, ADA went from zero because of

15 their ADA deficiency to, in fact, slightly above the

16 normal range for red cells from healthy donors. Their

17 bad metabolites, the deoxyadenosine metabolites dropped

18 down. When these patients initially present untreated,

19 these dAXPs are in the 50 percent or higher. And then

20 you can see that they also had immune reconstitutions,

21 so their T cell numbers and B cell numbers came up

www.transcriptionetc.com

245

1 after the gene therapy, after we stopped their enzyme

2 therapy. Next slide.

3 And so the second part of the slow road to

4 lenti -- so I sort of showed you the first half of

5 this. And so at the bottom half, so I just talked

6 about the Phase 1 and 2 trial. In fact, we decided we

7 should move to producing the cells in a cryopreserved

8 formulation and planned that as we then -- as this

9 property was licensed from our university, UCLA, to

10 this company, Orchard Therapeutics. So all this -- and

11 I moved into a new world of commercialization. And

12 they've then taken the ball from that point, and I will

13 be submitting a BLA application for licensure. Next

14 slide.

15 And so then, this slide just shows sort of for

16 this disease kind of the timeline from discovery of the

17 cause to treatments. And so ADA SCID was observed sort

18 of serendipitously being present in a few babies with

19 ADA SCID by Eloise Giblett back in 1972. Then we moved

20 forward to identifying the gene, cloning it, making

21 vectors and then a series of clinical trials. And in

www.transcriptionetc.com

246

1 fact, a gamma retroviral vector for this disease is

2 approved in the European Union as a drug called

3 Strimvelis which is available for therapy. And then

4 there's the lentiviral vector. So next slide.

5 So I showed this list before. I'll skip this

6 slide. Next one. So that's the work it took us to

7 develop a treatment for one of the hundreds of PIDs.

8 And in fact, I'm aware of currently that there are

9 three genotypes of SCID in gene therapy trials: ADA

10 SCID, X-linked SCID, and Artemis SCID. One form of

11 chronic granulomatous disease, Wiskott-Aldrich Syndrome

12 and Leukocyte Adhesion Deficiency, I believe those are

13 all the immune deficiencies currently being treated by

14 gene therapy, but in fact there's many others. So

15 there's another at least 17 other genotypes of SCID,

16 four other genotypes of CGD. And there's a whole list

17 of other even rarer monogenic primary immune

18 deficiencies that could be treated by this approach.

19 So the question is what would be needed to

20 develop individualized therapies for these other even

21 rarer primary immune deficiencies using an analogous

www.transcriptionetc.com

247

1 approach? Obviously, it's, you know, we can't spend 10

2 years, and companies are not going to invest in

3 diseases that are going to be treating, you know, two,

4 three patients a year. And then beyond the immune

5 deficiencies, there's all the other blood cell diseases

6 that would fall under this treatment: red blood cells,

7 white cells, platelets, stem cells, and then, you know,

8 even beyond that, other genetic diseases. Next slide.

9 So I guess the question is, you know, how do

10 we do this in less than 45 years? I mean, that's

11 obviously intolerable for patients who have diseases

12 that need to be treated right away or very soon. And

13 so over this period that I just showed you, the

14 development of this treatment, the investigative

15 capacity of biomedicine has vastly expanded as we've

16 all witnessed.

17 So we have far greater resources now,

18 reagents, materials, all the multiple -omics and

19 informatics to really move things quicker. And in

20 fact, we now have established a number of broad gene

21 manipulation capabilities both virally, vector addition

www.transcriptionetc.com

248

1 of AAV or lentis, and all the editing approaches that

2 Keith talked about briefly with growing positive

3 experience for safety and efficacy.

4 I think, you know, when this started out when

5 I -- at the beginning of the field, there was, you

6 know, it was unknown what kind of problems might

7 develop. And I think we are -- although there have

8 been some problems certainly along the way, we are

9 developing a growing experience of safety and efficacy.

10 And so we now have this cumulative experience in gene

11 and cell therapy product development, pre-clinical

12 evaluation, manufacturing, and clinical trial

13 performance. Next slide.

14 So how would newly identified genetic

15 disorders be fast tracked for individualized therapies?

16 Well, as someone said earlier this morning, the first

17 step is to understand the pathogenesis so we can

18 understand the therapeutic approach. One issue, you

19 know, is it an absent gene product that we just need to

20 add back the gene, or is there an abnormal dominant

21 negative or dominant adverse gene product that needs to

www.transcriptionetc.com

249

1 be overridden or knocked out?

2 And then we need to understand, you know, what

3 are the relevant cell targets? So if it affects the

4 blood cells, then what I was talking about would be do

5 allogeneic stem cell transplant or ex vivo gene therapy

6 may be beneficial. Other disorders like CNS, the

7 defective microglia, that are -- many of which are

8 blood cell derived can benefit. But this won't help

9 all the other organs most likely.

10 If it affects -- is an autosomal recessive

11 disease affecting motor neurons, then IV or intrathecal

12 routes might be needed. If it's an autosomal dominant

13 disorder affecting neurons, we may need to deliver the

14 cells or genes in situ. Deficiency of serum proteins

15 made in the liver, then intravenous AAV looks like a

16 very viable approach to treat those. And so there

17 won't be one size fit approach for all these genetic

18 diseases. It's really going to depend on which organs

19 are involved and what's the nature of the defect.

20 So we have a number of models to sort of work

21 this up in, and one of the most important always is

www.transcriptionetc.com

250

1 patient derived cells that have the defect that can be

2 studied. And now, we have great capabilities to make

3 induced pluripotent stem cells and make organoids that

4 can recreate elements of the disease. But, of course,

5 all the murine knockout and gene manipulation models

6 provide a way to test the new therapeutics.

7 And then, you know, once we've developed it,

8 we need to define the nature of it and, you know, is it

9 something that we've experience with? So is it just

10 AAV for a new genotype of a retinal disorder, for

11 example? Is it a cell type we've used, or is it

12 something new, and how much experience to do have also

13 then with the cell type hematopoietic stem cells, T

14 cells, liver cells, et cetera? Next slide.

15 So what are some of our opportunities to try

16 and use this experience for other diseases? Well, one

17 is that we can rapidly identify these responsible rare

18 gene defects underlying the inherited and de novo

19 diseases, whole exon, whole genome, CGH, et cetera. We

20 can relatively rapidly develop vectors and CRISPR-based

21 therapeutic targeting reagents. You know, within a few

www.transcriptionetc.com

251

1 weeks to months, we can have reagents targeted to a

2 specific disorder.

3 And as I said, the record certainly for lenti

4 and AAV vectors are mature with expanding safety

5 records. So it raises the question of how much pre-

6 clinical testing is needed if you just change a

7 transgene or you just change a small guide RNA. There

8 are clearly potential adverse events from a different

9 transgene or a different guide, but the more we can

10 leverage platforms and experience, the quicker it will

11 be. And then we can use gene engineered murine human

12 iPSC as I mentioned in patient derived cells to

13 determine the disease modifying activity. Next slide.

14 So some of the challenges still though are

15 quite significant. And one of them that has plagued

16 gene therapy and cell therapy since the beginning is

17 the challenge of in vivo delivery. So in vivo

18 delivering cells, genes or editing reagents to specific

19 cell types, and sites and tissues remains really

20 suboptimal in many cases. And I think that's still

21 limiting and, you know, I think is a very important

www.transcriptionetc.com

252

1 area. We're all hoping that there will nano techno- -

2 - nanoparticle technologies that will do that, but

3 that's still an early area I'd say.

4 And then beyond sort of the simple monogenic

5 disorders that I've talked about is there are many

6 other more complex genetic disorders that are

7 chromosomal deletions or duplications. Those are going

8 to be much more challenging to treat by either

9 replacing the large deletions or selectively removing

10 duplicate segments than are these single gene targets.

11 And then many of our diseases obviously are multigenic,

12 and these would be much more complex to approach by

13 either gene addition or editing methods. And these

14 cases, cell therapies that have the whole package might

15 be better.

16 And so I think Dr. Marks in his opening

17 remarks talked about a four to eight year typical

18 timeline, and I think I would agree with that. And so

19 the breadth of activity in toxicological testing for

20 pre-clinical studies that are typically done to support

21 IND are expensive and take a long time. And I was

www.transcriptionetc.com

253

1 probably low-balling at a $500,000 to $20 million and

2 one to three years. And I will stop there and look

3 forward to the discussion. Thank you.

4

5 PANEL SESSION WITH Q&A

6

7 DR. REINDEL: I'd like to invite our panelists

8 up to the front here, please. So in addition to Dr.

9 Kohn, who will continue to participate in the panel

10 discussion over the phone, we also have several FDA

11 representatives, including Dr. Lapteva from the

12 Division of Clinical Evaluation Pharmacology and

13 Toxicology and the Office of Tissues and Advanced

14 Therapies and Dr. Xu, who is a Senior Mathematical

15 Statistician in the Office of Biostatistics and

16 Epidemiology.

17 So I see no one has approached the microphones

18 yet. I'd like to encourage the audience to do so. If

19 you have questions, we'd really like to engage in

20 discussions, but I'll be happy to start the discussion

21 off with a question that I think is relevant to both

www.transcriptionetc.com

254

1 Dr. Kohn and Dr. Schooley. I was really impressed with

2 the way that both of you sort of united resources

3 across the country to enable collaboration in a space

4 that -- where you may be able to only enroll one or two

5 patients with a certain condition. Can either or both

6 of you talk a little bit about strategies that have

7 been effective to promote that kind of collaboration?

8 DR. SCHOOLEY: I think the main thing is

9 really being open to collaboration and realizing we're

10 all facing the same problems there. We gain by

11 collaborating. And there has been a lot of publicity

12 about MDR infections, so patients drive a lot of this

13 as well. And I think listening to patients and

14 physicians and their needs and trying to meet them is

15 one of the things we should do as investigators. I'm

16 sure that -- let's move to a genetic perspective on

17 that as well.

18 DR. KOHN: Yeah. And if I can comment, so

19 we've done a few trials for ADA SCID and CGD, for

20 example, with two or three or four different academic

21 sites. And besides having the colleagues to

www.transcriptionetc.com

255

1 collaborate with, which is enjoyable, spreading it out

2 geographically is helpful for patients so they don't

3 all have to go to one place in the country. And one of

4 the things that we've learned that's very important,

5 obviously, is having very careful monitoring of the

6 sites so that everyone is doing things exactly the

7 same.

8 And obviously, drug companies do this all the

9 time when they're doing multi-centered trials. But

10 coming from the academic perspective, it's a lesson we

11 had to learn to have ongoing active monitoring of the

12 clinical data, and also, in our trials at least, we've

13 mainly been doing cell manufacturing at each academic's

14 GMP site. And to harmonize that activity takes a lot

15 of work because, in general, people think they all know

16 how to do it. And -- but if you work with sites that

17 are agreeable, you can sort of standardize even

18 something as relatively complex as processing

19 hematopoietic stem cells with viral vectors.

20 MS. MCGRATH: Yeah. Lynne McGrath. I was

21 curious about your comment that you're hoping that it

www.transcriptionetc.com

256

1 doesn't take 45 years. And I'd like to turn the

2 question on to the group to say that was a technology

3 that has evolved over the last 45 years, and I've

4 personally been involved with programs that took over

5 20 years. But the question is, when new technology

6 emerges today, how do we not let that go 45 years?

7 Because certainly we have a lot of things that

8 have happened. But just your thoughts on some of the

9 new and emerging scientific discoveries that -- how do

10 we get that baseline information to be able to use

11 those as therapies? You know, the 45 years may still

12 be -- hopefully not but may still be something that we

13 would consider because of, you know, going forward with

14 new discoveries. So that's kind of a question that I

15 have is how do we shorten that? I don't know if

16 anybody has any thoughts.

17 DR. KOHN: Well, I mean, one example are the

18 use of CRISPR. So you know, that was only really

19 identified, what, seven or eight years ago or

20 something. And they're already -- it's already in

21 clinical trials. And so clearly, we have accelerated

www.transcriptionetc.com

257

1 our process of -- from discovery to clinical

2 applications.

3 It'll still need to go through now the process

4 of Phase 1 and Phase 2 trials to some extent. But I

5 think it's such a nimble platform for developing a

6 therapeutic if you're targeting a gene that I, you know

7 -- I think that will be one thing that will, for

8 genetic diseases at least, really accelerate the

9 timeline.

10 DR. SCHOOLEY: You could argue that phage

11 therapeutics has been going on for 100 years, so that

12 45 years is nothing. And I think the real key is

13 trying to understand what you're doing in as precise a

14 way as you can so you learn from it and can generalize,

15 and other people can either repeat or improve on the

16 experience. Where we make mistakes is where we do

17 things without characterizing them as carefully as we

18 can to be able to learn when it doesn't work, to learn

19 from things when they do work, and to build on what we

20 know.

21 So I think that should be probably the most

www.transcriptionetc.com

258

1 critical aspect of trying to accelerate discovery

2 across all fields. And I think it fits what was said

3 earlier today by some of the other people talking about

4 platforms that different companies might be developing

5 for different applications and indications. Why not

6 learn from each other? You're not competing for the

7 same product, and the more you share the more you can

8 focus on things that matter which is your particular

9 application.

10 So I think that phage therapeutics are the

11 same way. Companies aren't competing with each other.

12 It's not like people are lining up to decide which

13 company to invest in. People are trying to decide

14 whether this is crazy or not, and so the more

15 collaboration the better.

16 MS. WITKOWSKY: Hi, this is Lea Witkowsky from

17 IGI at UCSF and UC Berkeley. Hello, Dr. Kohn online.

18 DR. KOHN: Pleasure.

19 MS. WITKOWSKY: I'm wondering, all this talk

20 about the ability to separate product specific

21 attributes and processes from platforms seems like a

www.transcriptionetc.com

259

1 really important distinction, if we can come up with

2 ways to streamline creating evidence and being able to

3 leverage that evidence from one application to the

4 next. And since we're mentioning genome editing, I'm

5 wondering, Dr. Kohn in particular, as you've worked in

6 -- with hematopoietic stem cells across various

7 different diseases, how much do you expect an organ

8 system -- so for example, platforms of using lentivirus

9 in one organ system or tissue system to be

10 standardizable so that, if you're doing genome editing

11 for example, you're simply changing one component, kind

12 of leveraging the modularity aspect of things like

13 genome editing? I wonder if you could speak to that as

14 you've worked on multiple different diseases within the

15 blood organ. Thank you.

16 DR. KOHN: Yeah. No. Thank -- that's a

17 really good question. You know, I don't know fully the

18 answer. So if we use the example of using CRISPR-Cas9

19 to modify specific sites, obviously every time you go

20 to a new guide, it has a different on-target, off-

21 target profile. But as to -- by what Keith talked

www.transcriptionetc.com

260

1 about, our development to really identify that, you

2 know, that may be some -- you know, that kind of safety

3 analysis may be enough to let you not have to do the

4 full developmental package, just to target a different

5 genomic site, for example.

6 And the same thing with lentiviral vectors.

7 When you look at all the different papers now that have

8 been published looking at integration sites for

9 lentiviral vectors with a number of different diseases,

10 it's getting very monotonous because you kind of see

11 the exact same pattern. So we know that, and so it

12 then just becomes what are the transgene specific

13 issues that need to be studied. Obviously, you need to

14 show disease activity modification.

15 But beyond that can we, based on the class of

16 gene -- that it's a metabolic enzyme for example -- not

17 have to do all the extensive testings I showed you that

18 we did in mouse models, transplant models, et cetera.

19 And so I think some of it -- I think some of that is a

20 regulatory issue of what will be acceptable to allow

21 existing data to be leveraged and not have to start at

www.transcriptionetc.com

261

1 square one every time you change a guide or a cDNA.

2 DR. REINDEL: We have some questions from the

3 online audience.

4 THE OPERATOR: So we have two questions from

5 online. They're both pretty short, so I'll give them

6 to you at once. The first one is Dr. Schooley

7 mentioned about how phages are similar to antibiotics.

8 How do we ensure the environmental safety of phages?

9 Do we have any knowledge of how they might affect the

10 safety of the microbiome outside the patient? That's

11 question number one. And question number two is how

12 can the phage and gene therapy fields learn from each

13 other?

14 DR. SCHOOLEY: Well, question number one is

15 we're all loaded with phages, and we have -- we walk

16 around with more phages than we give to patients.

17 They've been around for 300 million years, and when

18 there is not substrate for them to grow in, they no

19 longer propagate. So phages are kind of self-renewing

20 and self-extinguishing when their substrate is gone.

21 One of the things that make them, I think,

www.transcriptionetc.com

262

1 less dangerous environmentally than antibiotics is

2 their spectrum is so narrow. So although I certainly

3 understand the issue, unless we were to come up with

4 some genetically engineered phage that had broad host

5 range, would take out all but one particular bacterium,

6 and that were, for example, Pasteurella pestis, I think

7 competition among phages will take care of that. And

8 the other question was related to --

9 THE OPERATOR: To field learning from --

10 DR. SCHOOLEY: Oh, I'm sorry. Learning from

11 each other. So let's, Dr. Kohn, see if there's

12 anything to learn and vice versa. Sorry.

13 DR. KOHN: Well, yes. I mean, I think, again,

14 I tend to think that a lot of these types of novel

15 therapies emerge from academic medical centers. And we

16 all spent our time in medical school and not doing

17 manufacturing. So I think some of the CMC issues that

18 you touched on we've also needed to learn and develop.

19 So I think, although we're making a different product,

20 we're using similar processes. So I think this bit of

21 crosstalk is useful. And maybe this is a Gordon

www.transcriptionetc.com

263

1 Conference topic or something to have adjacent meetings

2 and some overlap and talk about developing these kinds

3 of therapeutics.

4 DR. SCHOOLEY: Some of the same issues in

5 terms of producing phage are ones that Dr. Kohn is

6 talking about because different academic labs are

7 producing phages of different types but are beginning

8 to converge in terms of how they're purified. And

9 those kinds of convergences, I think, are very helpful

10 and make it easier to think about what's being done in

11 different places as well.

12 DR. KOHN: Right. And standardization of

13 potency testing and even titering is something that, at

14 least for lentiviral vectors, is totally lab specific

15 what a titer value is because there is not standardized

16 method. So I think, again, we face a lot of the

17 similar issues in these products even though they're

18 different products.

19 DR. REINDEL: If there are no additional

20 comments from the audience, I had another question that

21 I think will be helpful to the audience to hear. Could

www.transcriptionetc.com

264

1 you, Dr. Schooley or Dr. Kohn, discuss any specific

2 challenges you've faced in the design of these clinical

3 trials in terms of the challenges that you discussed

4 just now in terms of product and the approaches you've

5 taken to overcome those challenges?

6 DR. SCHOOLEY: Well, I think the challenges

7 that are common to both are -- have to do with the fact

8 that we're -- that the interventions are quite

9 individualized from patient to patient. And we have to

10 think about how to, as one person put this morning,

11 talk about the process by which they are made and what

12 standards and what metrics are used to say that these

13 products are similar enough that, when you use them in

14 different patients directed at the same organism but

15 with a different, for example, host range, that you're

16 -- you can aggregate the data in a generalizable way.

17 So being able to characterize the products in

18 a way that you can talk about their potency, talk about

19 their host range, what receptors they use, things that

20 let you, again, know what you're giving makes it easier

21 to design a hypothesis generated trial. That looks at

www.transcriptionetc.com

265

1 both clinical endpoints and biology and to -- at the

2 same time you're looking at the clinical endpoints,

3 learn enough about whether you're delivering the --

4 just like you're delivering a gene to the cell you want

5 to get to, we have to deliver the phages to the site of

6 infection, know they stay there, know that they remain

7 active, and measure those things at the same time the

8 clinical trials are being designed.

9 So what the challenge is there is that many of

10 the companies that are developing phages are, as

11 therapeutic agents, are relatively thinly capitalized.

12 And it's very difficult for them to support a lot of

13 the translational research that needs to be done. So

14 finding ways to get that done at the same time, I

15 think, is critical to moving the field as a whole

16 forward. But I'll stop there because there are

17 obviously other things that have to do with oncology

18 that are important, too.

19 DR. KOHN: Right. Well, so of course number

20 one is always funding. So as I showed on the timeline,

21 we had a -- every six months to a year or so we had to

www.transcriptionetc.com

266

1 apply for another set of funding to make it to the next

2 step. So I think CERM in California has done a very

3 nice job of sort of laying out their funding mechanisms

4 to follow the developmental timeline. And I think some

5 of the NIH institutes are also moving towards sort of

6 more multi-stage funding, so you don't need to back to

7 complete new R01 application for each stage of the

8 product -- project. So that's one of the issues.

9 The timelines are long for developing these.

10 And for academic careers it's not the best thing if

11 it's going to take you, you know, 10 years from when

12 you start to when you have your Phase 1 trial done.

13 It's hard to become an associate professor if you start

14 as an assistant with that. And so that's a challenge.

15 And then, you know, it's -- they're expensive.

16 To manufacture these products at high quality costs a

17 lot of money for the GMP, for the testing, the staff,

18 and also then the clinical trials are expensive. So I

19 guess that comes back to my first point of funding.

20 And so, you know, I think those have been the

21 challenges. We've obviously overcome them because

www.transcriptionetc.com

267

1 we're talking today, but those, I think, probably limit

2 what's developed.

3 DR. SCHOOLEY: Stability of funding is really

4 critical in phage therapeutics as well. The -- we've

5 seen over the last -- this trial that we talked about

6 briefly today has been in the works for two-and-a-half

7 years and watched multiple companies come and go, each

8 of which has gone down because of inability to maintain

9 their development plan. So it's critical to have

10 overarching mechanisms and support approaches that let

11 you plan for something that be carried out from

12 beginning to end. And having the government support

13 some of the basic and translational work, really, I

14 think move both fields forward in a way that get us

15 products, which is what we're all trying to do.

16 Because that, at the end of the day, is why we go to

17 the lab.

18 DR. KOHN: Yeah. And just to comment -- and

19 these challenges are probably even more intense for

20 individualized therapies, to get back to the theme,

21 that, you know, a one-off therapy that might be

www.transcriptionetc.com

268

1 lifesaving, if it costs a lot, if it takes a long time,

2 won't be valued. And so we have to find ways to do

3 these quicker and cheaper. Not sure what that answer

4 is, but I think that's the challenge.

5 DR. LAPTEVA: Yeah. So I would like to make a

6 comment and perhaps address some of the questions that

7 were asked earlier about product specific versus

8 platform and how we approach individualized

9 therapeutics and how if we have only one patient that

10 would need to be treated with a particular therapy, how

11 can we make this clinical development program looking

12 efficient and really deliver to the patient who needs

13 the therapy? A number of people this morning and the

14 afternoon spoke about the need to digress, to some

15 extent, or maybe apply regulatory flexibility to the

16 traditional medical product development model in order

17 to make the development of individualized therapeutics

18 more efficient. And although at this stage we don't

19 know collectively how these development programs may

20 look like and it's likely there would no one size that

21 would fit all and some of them will be very different

www.transcriptionetc.com

269

1 from others, we could at least try to identify some the

2 factors that would influence this digression from the

3 traditional development.

4 So one potential factor that we've heard about

5 today, and I could foresee, is the ability to make a

6 reasonable prediction about the product effect at the

7 time when the decision to treat for therapeutic

8 purposes is made. Another is the general expectation

9 that individualized therapeutics should work in

10 patients for whom they've been designed. And one other

11 important aspect, I think, is the determination of the

12 dosing. This is one of the very challenging aspects in

13 the development of individualized therapeutics.

14 But speaking about the decision to treat, if

15 you look at the traditional model of product

16 development, the decision to treat the disease for

17 therapeutic purpose typically does not come into the

18 picture until later because when people participate in

19 clinical trials, and when it is a group setting where

20 some people are treated with the investigational

21 therapy -- and the drug is the same for everybody --

www.transcriptionetc.com

270

1 and some people may be treated with placebo, and some

2 may be treated with active comparator --it depends on

3 the clinical trial design -- there is a little more

4 acceptance from the perspective of the patient as well

5 as the investigator that the product may not work. It

6 didn't work. It was investigational. This was a

7 clinical trial which failed to demonstrate the product

8 effectiveness.

9 If the product does work and there are

10 appropriate statistical methodologies that support the

11 positive therapeutic effect of the product, then we can

12 make an inferential conclusion that the patient

13 population with the disease will likely be benefitting

14 from this product. So when the next patient with the

15 disease comes to their physician in clinical practice

16 and the decision to treat is being made, then both the

17 patient and the physician have already some information

18 that gives them the ability to reasonably predict the

19 treatment effect because you would know that patients

20 who had the same disease, maybe similar

21 characteristics, were treated in clinical trials. And

www.transcriptionetc.com

271

1 you have some ability to predict the magnitude of the

2 effect of the product, whether it would or would not

3 work, and to understand some basic toxicities.

4 With the development of individualized

5 therapeutics, the decision to treat comes much earlier.

6 Even at the investigational stage, there is this

7 expectation that the product should work because it was

8 designed for this -- for the particular patient. Yet

9 the step which is so common for all of the different

10 products about taking the representative sample of

11 patients, testing the hypothesis, observing the

12 effects, and then translating it to the population with

13 the disease is absent. So there has to be something

14 that fills the gap with individualized therapeutics.

15 It would serve the information -- as the information

16 with the sufficient predictive capacity to enable this

17 decision to treat.

18 And so speaking of the novel technologies,

19 what we see although in very, very few examples of such

20 individualized therapeutics developments -- and thank

21 you to the person who spoke this morning who is the

www.transcriptionetc.com

272

1 patient with cancer. You didn't realize probably, but

2 you were one of the examples of what we're actually

3 seeing. We're seeing development of new methods and

4 methodologies that are based specifically on that

5 individual patient's genetic parameters, physiological

6 parameters, understanding of the cellular metabolism,

7 how cellular phenotype may be changing with the

8 introduction of a transgene in that particular patient.

9 So what we will likely see in the field with the

10 development of individualized therapeutics is the

11 growth and development of these methodologies that are

12 predicting the individual patient's response to that

13 particular individual therapy. So that's one.

14 The other is the expectation that with these

15 types of treatments we will see positive treatment

16 effects, if not to say large treatment effects. So if

17 you take a gene therapy, for example, which is targeted

18 to correct a functional gene or if you take a cell

19 therapy that's intended to replace some lost functional

20 cellular tissue, then you would expect that not one and

21 not two, but many physiological processes, downstream

www.transcriptionetc.com

273

1 physiological processes will be affected. And likely

2 also maybe some anatomical changes will occur which

3 would result in previously unseen quantitative or

4 qualitative effects of the therapy previously unseen in

5 the disease progression.

6 And in that case, and my clinical colleagues

7 will understand me, something that we call minimally

8 clinically important difference will not be hinged

9 anymore on the comparison between the two groups. But

10 it would be very important to receive the input from

11 the patients and their caregivers. And that's where

12 the collaboration between the patient community and the

13 investigators would be very critical in understanding

14 as to what is the meaningful effect of that individual

15 therapy or a number of individual therapies and not

16 only that but also how these effects progress over time

17 and how we can monitor and evaluate the effects of

18 individual treatments in the long term.

19 And we have to create systems to do this. And

20 I'm sure the next panel will be talking about it --

21 systems that are able to collect clinical data. But

www.transcriptionetc.com

274

1 not only that, but to go back and maybe -- somebody

2 also mentioned it this morning. If you have a

3 predictive statistical model which feeds and includes

4 physiological variables and IT variables and other

5 variables, why can't you fit -- feed the clinical data

6 back into this model to make it a little more

7 predictive? And so this is something that we will also

8 likely see developing.

9 And lastly, for collection of safety data,

10 particularly when it is a platform based product where,

11 say, a vector treatment that's been optimized already

12 with understanding how different elements of the vector

13 may interact with one or more transgenes that would

14 potentially be inserted for treating different diseases

15 -- if you take this type of platform and we have a

16 clinical data collection attached to it and you may

17 call it a master protocol or a platform protocol -- but

18 it would be important to incorporate common variables

19 into that platform to enable perhaps a meta analytical

20 activities to be done later down the road specifically

21 with regard to safety because safety could potentially

www.transcriptionetc.com

275

1 be evaluated across different diseases for very related

2 but slightly changed products.

3 DR. REINDEL: Okay. I think that concludes

4 the panel discussion for today. Thank you for all of

5 our -- to all of our participants.

6 DR. KOHN: Thank you.

7 DR. RAYCHAUDHURI: So, I'd like to thank all

8 the speakers and panelists and the audience for sharing

9 your perspectives. And I'd like to thank Dr. Reindel

10 for moderating the session. And Dr. Kohn, I'm very

11 sorry about your travel challenges but so very happy

12 that you could give your presentation and join the

13 discussion also. So we're going to --

14 DR. KOHN: Thanks for having me, Gopa.

15 DR. RAYCHAUDHURI: Thanks so much. So we will

16 take a short 10-minute break. And we have three

17 excellent speakers coming up in session four in what I

18 know will be a very thoughtful and very thought-

19 provoking session. So please take a quick break and

20 join us in ten minutes. Thanks.

21 [BREAK]

www.transcriptionetc.com

276

1

2 SESSION 4: PRODUCTS TO PATIENTS

3 DR. RAYCHAUDHURI: Okay. If everybody could

4 take your seats, we'd like to get started with session

5 four. So I just want to say, in addition to everybody

6 in the room we've had -- we have over 400 people online

7 who are following the workshop. So it's great to see

8 this level of interest.

9 So it's my pleasure to introduce Dr. Celia

10 Witten. Dr. Witten is the Deputy Director of CBER, and

11 she will be moderating session four, which focuses on

12 how to get products to patients in a timely manner and

13 maintain access for patients in a sustainable way. Dr.

14 Witten.

15

16 SESSION 4 MODERATOR INTRODUCTION: DR. CELIA WITTEN

17

18 DR. WITTEN: Thank you. In the prior three

19 sessions, we talked about the scientific and clinical

www.transcriptionetc.com

277

1 aspects of development and heard about the importance

2 of regulatory flexibility leveraging knowledge across

3 applications, the challenges in manufacturing and

4 testing, and the need for collaborations. There will

5 be three speakers in this session: Jill Wood from

6 Phoenix Nest, Dr. Alison Bateman-House from New York

7 University Langone Health, and Dr. Phillip Brooks from

8 NIH. For the panel discussion, the speakers will be

9 joined by Captain Julie Vaillancourt from the Rare

10 Disease Program at CBER and Dr. Chip Schooley who

11 participated in the prior session.

12 The focus of this session is on ethical

13 issues, collaborations, and stakeholder roles in the

14 end-to-end development of individualized therapeutics.

15 In general, the role of individual stakeholders can be

16 quite different in the development of products for rare

17 diseases than in the development of products for common

18 disease indications. This may be even more so in the

19 development of individualized therapeutics intended for

20 one or a small number of individuals.

21 As stakeholders take on certain roles in the

www.transcriptionetc.com

278

1 development of these products, including close

2 collaboration with other stakeholders, certain ethical

3 issues arise. Furthermore, the focus of collaborations

4 may need to go beyond development to consider future

5 sustainability issues, since not all products or

6 approaches may be commercializable. In addition, new

7 development paradigms may pose ethical conundrums that

8 are not necessarily features of the standard drug

9 development paradigm. Therefore, these three issues

10 are somewhat related.

11 Some of the ethical issues that we see are

12 noted on this slide. One question that needs to be

13 addressed for any drug product and development is the

14 determination of sufficient manufacturing and safety

15 information for a trial to proceed and also, as we've

16 heard, sufficient information to develop other aspects

17 of the trial, such as determining the starting dose for

18 a product that can only dosed once. So these kinds of

19 questions about manufacturing safety information may be

20 even more of an issue for products for a single

21 individual or a small number of patients because

www.transcriptionetc.com

279

1 testing can be resource intensive.

2 Note that the determination of whether there's

3 sufficient information for a trial to begin is not a

4 simple risk-benefit question because there may be

5 considerable uncertainty in the safety testing or, for

6 that matter, an assessment of activity. Often the

7 discussion regarding N of 1 development is focused on

8 the acceptability of administering the product to the

9 individual. But the boundary between research and

10 clinical care may be very unclear. However, we need to

11 make sure we learn from each clinical investigation,

12 including single subjects.

13 Some of the development programs have been

14 funded or championed by a patient, family member, or a

15 small group of patient families. While this is

16 commendable and there have been some striking

17 successes, it involves a heroic effort on the part of

18 these family members, many of whom describe the effort

19 as being equal to more than a full-time job. Do we

20 need to figure out what to do to ensure that this is

21 not the expectation for families and caregivers? And

www.transcriptionetc.com

280

1 that is one of the issues regarding funding of

2 development. Another potential ethical issue,

3 particularly with regard to funding, is the question of

4 how development decisions are made, for example, which

5 patients get treated. Dr. Bateman-House will be

6 providing her perspective on ethical issues that arise

7 in this area.

8 Patients and patient groups have always been

9 stakeholders in medical product development, but these

10 groups are increasingly taking a lead role,

11 particularly for products with rare diseases. We will

12 hear about an outstanding example of these efforts from

13 Jill Wood in this session. Academic developers play a

14 larger role in the development of many of these

15 therapies than traditional pharmaceutical companies, in

16 many cases developing and performing early clinical

17 testing and, in some cases, partnering with

18 pharmaceutical companies or forming small companies to

19 shepherd the product across the finish line.

20 Philanthropic organizations are playing an increasingly

21 significant role in development programs for some

www.transcriptionetc.com

281

1 pharmaceuticals and will be important in this area

2 also.

3 We've already heard from a number of speakers

4 that collaborations are needed. These collaborations

5 are needed for many reasons, which have been discussed

6 in previous sessions. Some of the products may have

7 limited commercial viability. Academic developers may

8 have limited resources. In addition, as previous

9 speakers have noted, information sharing to eliminate

10 duplication of expensive development work would help

11 these products move forward.

12 There are many different models for

13 collaborations, and some amazing individuals and

14 organizations are currently leading collaborations to

15 develop individualized therapeutics. And there are

16 many models for information sharing. We can learn from

17 these examples as we work together to find a way to

18 develop and make available such products in a way that

19 is ethical and as resource efficient as possible.

20 However, it's possible some new models of

21 collaborations are needed because development work may

www.transcriptionetc.com

282

1 be duplicated, and these innovative areas benefit from

2 information sharing. There may be something to be

3 learned across patients for each product platform, as

4 we've heard during the prior sessions. And this could

5 include what we learn for a gene therapy vector for

6 related applications that could be related by tissue

7 target or by disease.

8 There could be a bio-distribution study done

9 for a vector that there'll be something gained in our

10 knowledge for applications with similar tissue targets

11 and delivery. And we may learn more about test methods

12 that will help in product development by gaining an

13 understanding of how these test methods work in

14 different applications. So it's important not just to

15 think about how to develop the therapy for each

16 individual or small group of patients but how we can --

17 you have to scroll down. Can you scroll down? -- how

18 we can do that sustainably across a range of

19 applications so valuable information is not lost or

20 overlooked but can be added to. Thus, we need to think

21 about development more holistically than one patient at

www.transcriptionetc.com

283

1 a time when we can. And we're going to hear a great

2 example of an effort in that direction from the talk

3 that P.J. Brooks will be giving during the session.

4 One last point I want to mention, we've been

5 referring to sustainability in this workshop. Why is

6 this important and why do we mention it when we discuss

7 collaborations? The questions of how to sustainably

8 provide products is important because, once a product

9 or a platform for designing individualized products has

10 been developed, how will patients be able to receive

11 these products? The traditional model has been for

12 pharmaceutical companies to take over production and

13 delivery. But some of these products may be valuable

14 to patients but not necessarily commercializeable.

15 Next slide.

16 So you've seen versions of this slide in each

17 of the previous talks illustrating the challenges and

18 opportunities in this -- in the area of product

19 development. This slide illustrates the fact that

20 traditional roles of how products are developed from

21 discovery through marketing are being upended, with

www.transcriptionetc.com

284

1 discovery being driven by patients and advocacy groups

2 as well as by NIH and other research grants funding or

3 by pharmaceutical companies. This upending of

4 traditional roles is seen all the way through

5 development. The question is where are the

6 opportunities for stakeholder collaboration among these

7 stakeholders and end-to-end development of

8 individualized therapeutics so we can move the field

9 forward?

10 Examples of areas for collaboration can

11 include availability of GMP grade material for clinical

12 studies. We have heard, both at this meeting and at

13 other venues held, how this challenge is limiting

14 development of AAV vector-based gene therapy. If this

15 same GMP grade material were available across multiple

16 researchers, for each product and development across

17 researchers, shared safety testing information could

18 help to reduce development costs for each product in

19 development for each researcher.

20 In addition, shared clinical data for

21 leveraging understanding would become possible.

www.transcriptionetc.com

285

1 Development of templates to facilitate IND submission

2 for collaborative development program might involve

3 multiple products but also could streamline

4 development. So as I mentioned, Dr. Brooks in this

5 session is going to describe NIH efforts of a

6 collaboration that's aimed at addressing these issues.

7 There are challenges for collaborations, also.

8 I've only listed a few examples on this slide, and I'm

9 sure the audience can come up with many more. But

10 including -- included among the examples would be

11 funding and governance. And by governance, I mean how

12 decisions are made regarding the collaboration goals

13 and the process, including the kinds of development

14 decisions I mentioned earlier when I discussed ethical

15 issues. And then there's the question of how

16 intellectual property is treated which also has come up

17 in this meeting.

18 And the last item, sustainability, I've

19 already mentioned, which is that we have to think about

20 not just development but what happens after the

21 development is completed because the goal of all of our

www.transcriptionetc.com

286

1 efforts should be to make sure our patients are able to

2 benefit from these new development programs. And

3 therefore, thoughts about how to do technology transfer

4 for commercialization are important, and also how to

5 ensure continuing availability.

6 I summarized at a high level the background

7 for the three related issues in this session: ethical

8 issues, collaborations, and stakeholder roles. And now

9 I'd like to introduce the first speaker. Do you have

10 the -- So our first speaker is Jill Wood. She's the

11 co-founder of Phoenix Nest, Inc., and she also has

12 funded a foundation to look at -- to try to develop

13 natural history studies and to try to develop funding

14 for Sanfilippo Syndrome. So please welcome Jill Wood.

15

16 THE TRIALS AND TRIBULATIONS OF DRIVING A TREATMENT FOR

17 AN UBER-RARE DISEASE TO THE CLINIC AND BEYOND-A

18 PARENT’S PERSPECTIVE - MS. JILL A. WOOD

19

20 MS. WOOD: Hi. Thanks for having me here. I

21 am Jill Wood. I am the mother of a child with an uber-

www.transcriptionetc.com

287

1 rare disease called Sanfilippo Syndrome Type C. And

2 I'm gonna walk you through our journey of what it's

3 been like trying to create a treatment and bring it to

4 the clinic for this disease. I really want to thank

5 the FDA for having me here today. It means a lot to

6 me. I greatly appreciate the fact that the FDA is

7 starting to look at the issues coming from the families

8 that are driving the science here.

9 So my clicker -- so before we can get to our

10 gene therapy utopia, we need a need. My need was born

11 on July 30, 2008. Jonah was full term. He was

12 absolutely perfect. We had no idea that he harbored

13 this insidious disease.

14 It was at our first year well visit that our

15 pediatrician noted that Jonah's head circumference was

16 off the charts, and we should probably go and get it

17 checked out. So we did. With due diligence, we went

18 to get our MRI, which was done at NYU, very fortunate

19 that we landed at NYU where the lab technicians there

20 knew exactly what they were looking at, which is

21 extremely rare in a disease like this. So they

www.transcriptionetc.com

288

1 suggested to our geneticist that we do a panel screen

2 for the MPSs.

3 So real quick about the diseases, I don't want

4 to bore everybody with any more science. Sanfilippo

5 Syndrome is MPS III. There are several different

6 MPSs. You've probably heard of many of them. The top

7 portion of those MPSs all have treatments. Sanfilippo

8 is the only version that does not have a treatment.

9 And I have four sub-types here: Type A, B, C,

10 and D. My son has type C, and my company is focused on

11 a treatment for type C and type D. And you can see

12 that our diseases are 1 in 1.5 million, which estimate,

13 maybe there's 100 kids in the United States by that

14 number. But I only know of 20 for type C and four for

15 type D.

16 Because our disease was so rare, we were told

17 that nobody was picking us up, that nobody would touch

18 us. One venture capitalist actually told me he

19 wouldn't touch my disease with a 10-foot pole. Yeah.

20 So you know, this was my first child. My husband and I

21 had done everything right. We had bought our first

www.transcriptionetc.com

289

1 house, paid off our school loans, and here you're gonna

2 tell me that my child has a terminal illness and

3 there's nothing I can do about it.

4 We hit the ground running, and we called

5 people that cared. We found these guys off of PubMed.

6 We brought our physicians -- fortunately, again, I'm

7 from Brooklyn, and I'm surrounded by wonderful

8 hospitals with geneticists and neurologists that were

9 ready to jump on our bandwagon and help us out. We had

10 a meeting in 2011, and we sat down with a few patients

11 that I had found, and our physicians and our

12 scientists. And we hammered out what it was that we

13 were going to do and that was to go for gene therapy.

14 So I'm going to go through -- the FDA asked me

15 how hard it was, you know, what did I have to do and

16 how we could do it better And so my first learning

17 curve was working with academia and finding the

18 scientists to help you start your program, getting the

19 -- your mouse model made. There's Alexi Bedeski with

20 our first mouse. I asked him to name him Juniper

21 because I wanted to name my second child Juniper. But

www.transcriptionetc.com

290

1 I'm not gonna ever have any more children, so there's a

2 Juniper.

3 But all of our science was funded outside of

4 the United States. It began in Montreal, with Brian

5 Bigger up there, and in Manchester. We funded these

6 guys through grass roots fundraising. We nickeled and

7 dimed it. That's a garage sale, a picture of a garage

8 sale there. This is how we did it.

9 It was a learning curve for me. We had to

10 write grants, hired lawyers, made sure that our

11 scientists were held accountable, that we had

12 milestones, and their payments were conditional on

13 their milestones. But for the people on the phone,

14 it's hard working with academia. These guys have a

15 school schedule. They take a lot of time off. And

16 sometimes they're post-docs, graduate, or they want to

17 get married or something. And you have to hire someone

18 else. It's not easy.

19 So during this process of nickel and diming

20 and trying to find -- scraping every dollar that you

21 possibly can, I met a gentleman who suggested that I

www.transcriptionetc.com

291

1 create my own company and go for NIH small business

2 grants. And I was like, you know, that sounds like a

3 great idea. Let's go for it. So we applied for an

4 STTR, these small technology transfer grants. I hope

5 many of you know who they are, what they are.

6 And unfortunately, there's small business

7 technology grants, and they don't want to fund

8 researchers that don't live in the United States.

9 Makes sense. So my type C research has not yet been

10 funded, but during this time I created a knockout mouse

11 for MPS III D. I actually applied for a competition,

12 Assay Depot, I want to give them some props for this Be

13 HEARD contests. I applied and we won the main prize, a

14 knockout mouse.

15 And that was 10 years ago about -- no, maybe

16 eight years ago, and that was like winning a car. You

17 know, it was before CRISPR, so that was pretty amazing.

18 Then, I went back, and I licensed our gene therapy

19 program from Manchester, brought it back into the

20 United States, and we're now working with our dosage

21 study with a CRO here. And I'm being helped with

www.transcriptionetc.com

292

1 additional funding from our friends at the Cure

2 Sanfilippo Foundation. Sorry.

3 So where we did not do so well with type C

4 grants, we have excelled with III D grants. And again,

5 major props to the NIH, we are now well on our way to

6 the clinic for MPS III D with an enzyme replacement

7 therapy. That one -- that started off from winning

8 that knockout mouse and snowballed into almost $7

9 million in SBIR grants. And like I said, there's four

10 patients, and those come from two different families, a

11 set of identical twins that you can see here, and a

12 younger family as well.

13 So while I'm developing treatments for type C

14 and D, I'm watching my sister diseases Sanfilippo type

15 A and B, which are much more prevalent than type C and

16 D. And it was really very exciting. They had several

17 programs in the pipeline. then all of a sudden, look,

18 Alexion/Synogeva, their ERT in MPS III B was shelved.

19 It was shut down; the trial was shut down before it

20 even got to the end. Same goes for ERT for -- with

21 Shire. BioMarin, if you might've heard from the press

www.transcriptionetc.com

293

1 releases, they did divest and were found a partner,

2 Levits. So that, thankfully, that trial will continue

3 on. But here we're sitting in limbo with Sobi, who

4 also wants to divest their ERT for MPS III A. You can

5 imagine how devastating this is for our community.

6 This is Will. Will's parents were told the

7 same thing that I was, that their child had an ultra-

8 rare disease and there was no treatment. A few months

9 later, lo and behold, here's a trial, and Will was

10 accepted into it. It says Shire right there. I'm

11 sorry to beat on Shire. That was actual Alexion's

12 trial. So sorry, Shire. That was actually Alexion

13 that dropped that trial, and they shelved it. They

14 dropped Will cold turkey. Took him straight off of his

15 ERT and is back to being told, “I'm sorry, your child

16 is going to die.”

17 Why is this happening? There's a lot of

18 speculations. I, for one, think that they have chosen

19 wrong endpoints. They're looking for cognitive changes

20 when our children have profound brain damage. And

21 we're not gonna change the cognitive in our children,

www.transcriptionetc.com

294

1 and it's really not what we're looking for. Financial

2 risks, a lot of these companies as you can see, have

3 changed hands, and not everybody's on the same page as

4 the previous CEO. BioMarin, bless them -- have nothing

5 against BioMarin -- but they went in a different

6 direction. They're not doing ultra-rare diseases

7 anymore.

8 And then, I think people don't realize our

9 diseases are slow to progress, but Will, Jonah, they

10 could die in their sleep tonight. It just happens.

11 Did I turn off my slide? Okay. Oh, but that -- okay.

12 This slide just kind of goes to shows you how

13 much our patient community has done. I am very, very

14 proud to be part of this Sanfilippo community. I don't

15 know that I've ever seen another rare disease community

16 work as hard as we have. Again, there's four different

17 sub-types, and we have four different enzymes. And

18 there's some things that are different about us.

19 So take for example these six programs that

20 have gene therapy programs in the works right now. The

21 first one nationwide was entirely -- pre-clinic was

www.transcriptionetc.com

295

1 entirely paid for by these foundations. And I think I

2 forgot to write the Cure Sanfilippo Foundation up

3 there, too. But all these foundations came in and

4 funded the pre-clinical, and then Abeona came in and

5 licensed it. Right now, they have A and B in the

6 clinic, and hopefully, it'll stay. And we'll have our

7 first treatment for Sanfilippo.

8 Lysogene was started by Karen Aiach.

9 Unfortunately, her daughter Amelia passed away just

10 before November and did not benefit from this

11 treatment. The trial still goes on underneath Sarepta.

12 I think I heard somebody from Sarepta here, so thank

13 you Sarepta for picking this trial up.

14 And then we have Estevee out in Spain, lay

15 low. Notable, Amicus has picked up MPS III A and B.

16 No, John Crowley is not a Sanfilippo parent, but he is

17 a Pompeii parent. Orchard was entirely funded pre-

18 clinical from the U.K. MPS Society. And, I think it

19 was the Ormond Street Hospital as well. And then

20 there's Phoenix Nest, who's driving the science for

21 type C and D.

www.transcriptionetc.com

296

1 The patient organizations, just, I mean, does

2 this shock the people in the audience that this has

3 been funded by us? And it shouldn't be this way. I

4 mean, we have children at home that are dying, and

5 we're here working our butts off trying to create

6 treatments for the next generation.

7 Okay. Don't you love the emojis on Apple? So

8 and this is also -- I had to throw this in there

9 because this really annoys me. It keeps me up at

10 night. Our SBIR grants, you know, you have a

11 collaboration with academia, and they get subawards.

12 They get a nice substantial subaward. They don't --

13 pays for all their overhead.

14 And, you know, they get to ask for some, a

15 bioreactor, you know, a $300,000 bioreactor. They get

16 some nice equipment. And then they get licensing

17 rights because it happens on their -- in their hallways

18 and on their property. And then they get all the fame

19 and glory and get to write papers and publish and go

20 around.

21 But then, I have to go back and license what

www.transcriptionetc.com

297

1 it is that my company won. I have to go and license

2 this. And what I don't think people really realize is

3 that I have to hire a lawyer. I have to hire patent

4 lawyers. Do you guys have any idea how much lawyers

5 cost? SBIR grants do not cover lawyer fees.

6 So to add insult to injury, here we are just

7 trying to scrape by on a disease for four kids. I

8 spent -- I actually spent over a year fighting this.

9 No milestone payments, no upfront fees. I mean, it's

10 just ridiculous to ask that from me. When you do, they

11 don't realize that you're sucking any incentives that I

12 had for commercial partners away. When you're treating

13 four kids, how many patients -- how much money are you

14 gonna make off of this?

15 So I'm throwing that out there. If there's

16 something we can -- some template we could put on our

17 SBIR grants that -- what do I want to say -- makes both

18 sides happy but realizes that these are ultra-rare

19 diseases and you can't treat me like a Parkinson's drug

20 company. Oh, boy. Things that I think the FDA, or the

21 NIH, could do for us that are no brainers, and I know

www.transcriptionetc.com

298

1 you know that they're no brainers because we've talked

2 about them a lot, natural history studies. One good

3 one, one we started -- we knew we needed to do a

4 natural history study.

5 But my families did not want to fund a natural

6 history study. They are extremely expensive. And when

7 you have $1 million dollars, do you want to send it

8 sending your kid to a clinic to be poked and prodded,

9 or do you want to put the money in making a mouse model

10 and making a gene therapy? I mean it's the mentality

11 of it. The families want to fund the research. And

12 now we're stuck.

13 We really, really have got to get our natural

14 history study undergoing. Mouse models, I have them.

15 But there are so many rare disease groups that don't

16 even have mouse models. And they're extremely easy to

17 make now and to house. I think if there was something

18 that the FDA could do for us, it would be to go through

19 and find out who doesn't have their mouse models.

20 Registries, registry's another major

21 contention amongst our patient groups. It's something

www.transcriptionetc.com

299

1 that we absolutely have to have as well, but again,

2 they extremely expensive to maintain. You have to be

3 HIPAA approved, GDPR approved, and those things have to

4 happen yearly. And who's going to do that? Who's

5 going to keep up on that? We really need help managing

6 and maintaining that. And that's, I think, another

7 thing our federal government could do for us and could

8 do it flawlessly.

9 And then one last topic I wanted to sneak in

10 here because nobody really likes to talk to their

11 patients about this -- patient groups. But biobanking

12 -- oh, there is a major lack of donations out there.

13 And if we could have this sensitive conversation with

14 our families that it is imperative that we keep some of

15 these tissues, , eyes, so you know -- it would be

16 extremely helpful. I think we talked a lot about

17 mutations as well and knowing the mutations of these

18 families. And I like this company, and the NIH

19 supports them as well. And I'm putting that up there

20 so people can take notes at home.

21 Here comes our Orphan Drug Act. And we love

www.transcriptionetc.com

300

1 Abby Meyer and what she did for us. That was done

2 almost 25 years ago, and I think it's about time that

3 we update our Orphan Drug Act. We all know that there

4 are 7,000 rare diseases, and here we consider that 1 in

5 20,000 with a patient population of 200,000. That's a

6 rare disease. So if you're a drug company and you're

7 gonna create a drug for a rare disease, who are you

8 gonna pick? One with 100,000 patient population or a

9 patient population of 20?

10 We're starting to talk about ultra-rare quite

11 a bit, but what is that number? What does that look

12 like? How do you go from 200,000 to 100? So if you're

13 gonna -- I think we need to get ultra-rare on the map.

14 We might as well get uber-rare on the map as well. And

15 we really need to figure out ways that we can

16 incentivize this.

17 I've been doing this for nine years. My drugs

18 are ready to go to the clinic. We're handing

19 everything over on a silver platter, and I still do not

20 have anybody knocking on my door. Pediatric review

21 vouchers are absolutely amazing, but it's still not

www.transcriptionetc.com

301

1 trickling down to us.

2 Just to let you know, we did do our pre-IND

3 meetings for both type D and type C. They went very

4 well. We went in really early because we were scared

5 about the slowly progressive heterogeneity, the tiny

6 patient population, and the lack of natural history.

7 How were we going to pull this off?

8 And CDER and CBER both gave us the same advice

9 was get creative and get more natural history, which we

10 are trying. I'm trying so hard to get our natural

11 history off the ground. And we will be looking at

12 mosaic endpoints, kids being their own control. We're

13 hoping to hang our hat on the guidance that was put out

14 last June and using heparan sulfate as our surrogate

15 marker.

16 Okay. You don't have to stare at this long.

17 You can look over at me, but you know, you read what

18 Sanfilippo looks like on paper. You read what these

19 diseases look like on paper, but to see it for yourself

20 is a huge difference. Yes, this is my bathroom at 8:00

21 in the morning. “Mom, I had an accident.” Okay, I'm

www.transcriptionetc.com

302

1 going to go and clean up his pants. You know, you're

2 parents. Somebody has an accident in his pants you

3 might throw them in the garbage. I look at my husband

4 and I --"Can we just throw the toilet in the garbage?

5 I can't deal with this.” Imagine this at Starbucks.

6 Picture that. Okay. You're never going back to that

7 Starbucks again.

8 Some families, their children scream and cry

9 nonstop for months on end, and nobody can figure out

10 what it is. Is it neurological? Does my child have a

11 bladder infection? Is it tooth decay? What is going

12 on here? And it al- -- it seems to end up being

13 neurological.

14 But these kids, I mean, it's terrifying for

15 the families. They don't sleep for months on end, and

16 these kids are mobile. So the families have to put in

17 safe rooms and lock the doors and lock everything down,

18 and their kids just walk around the room with the

19 lights on, switching it on and off, turning the T.V. on

20 and off. They're up all night long. It's horrible.

21 Imagine that.

www.transcriptionetc.com

303

1 So anyhow, yeah. This goes back to the point

2 that we don't care about cognitive endpoints. We need

3 to take care of some of the serious issues that we live

4 with day in and day out that will make our lives

5 better. I threw this in there, and we talked a little

6 bit about the workshop that happened last month for the

7 expanding AAV manufacturing capacity.

8 And that happened here, as well. And if -- I

9 encourage you guys to go back and watch this

10 conference. And I bring this up as -- for the end-to-

11 end gene therapy. And this company, I was very much

12 impressed with this company, the Discovery Lab, which

13 is doing exactly what we're -- we want to see happen.

14 I am now in the process of creating our

15 vector, putting it all together. This is another huge

16 hurdle for me and mind blowing. Three different

17 plasmids made at three different CROs that all have to

18 come together at one place and, if one isn't working,

19 then it throws everything off and you have to go back.

20 I mean, the level of expertise needed here is huge.

21 It's very time sensitive and the error for margin is

www.transcriptionetc.com

304

1 huge. So I commend this group for making this happen.

2 I would love to see it trickle down to the FDA for the

3 ultra-rare diseases as well.

4 Again, this was from that last conference, the

5 AAV conference. Last month alone I had three new

6 families from diseases that I had never even heard of

7 before calling me up that found me from random places

8 and say, “Hey, I need help. This is what's happening

9 to my child,” and it sounds very similar to what's

10 happening to my child. Each one of those diseases

11 breaks down into several other different subtypes.

12 They're all conducive to gene therapy. Just pull out

13 the gene and put in the next one.

14 And finding these gene therapy scientists, I

15 hate to even put pictures of Steve Gray up there. I

16 haven’t even talk about him because I don't want to

17 share him. But we have got to have more Steve Grays

18 out there and how it is that we're going to train these

19 people and, if they are there, to come out of the

20 closet somehow. You know, reach out to the FDA or the

21 NIH or Global Genes or EveryLife Foundation and say,

www.transcriptionetc.com

305

1 “Hey, I want to help. I have a lab; I'm interested in

2 gene therapy.” And these families are ready to

3 fundraise for you.

4 And yes, to my rescue, Jude Samulski, you

5 might have heard of him. His company AskBio has spun

6 out for Viralgene, and they have created a nonprofit

7 company where they are holding suites for uber-rare

8 diseases for people like myself. And we talked about

9 the bottleneck, these guys are holding a suite for our

10 gene therapy. So thank you very much.

11 Now, I always have to throw in my newborn

12 screening because I think this is vitally important for

13 all of our children, whether we have treatments or not.

14 Ignorance is bliss. That's my baby, a few weeks old.

15 But knowledge is power. Like I mentioned that Jonah

16 was diagnosed very early, he is the youngest child

17 known to ever be diagnosed asymptomatic without an

18 older brother or sister.

19 Because of that, he had tubes put in his ears.

20 He was a year-and-a-half. I didn't know he couldn't

21 hear. Put the tubes in, it gushed out, and he was

www.transcriptionetc.com

306

1 pointing out airplanes the next day. I was like he

2 couldn't hear that it was an airplane. It breaks your

3 heart to know that your child -- he can say ball, and

4 mom, and dad, but he couldn't hear. He also had bad

5 sight, so we got glasses.

6 His behavior, you can imagine during the

7 formative years how important it is to be able to hear.

8 Not only does it help you read and sit still and

9 participate in class, but it takes away that

10 frustration and helps with the behavior. Sanfilippo is

11 strife with behavioral issues. The kids are very --

12 can be very aggressive. So I actually feel guilty that

13 my son is doing better than any other Sanfilippo child

14 that I have ever met, and I attribute that single

15 handedly to the fact that he had early intervention and

16 we caught his hearing before he was deaf.

17 So that brings me to a close, and I have some

18 people that I want to thank. And lots of people aren't

19 on there. I can't thank everybody enough. There have

20 been plenty of professionals that have helped me with

21 pro bono services. But most importantly, I want to

www.transcriptionetc.com

307

1 thank my families.

2 We're very diverse. I have friends all around

3 the world. We don't speak each other's languages, but

4 we're family. And if it wasn’t for them, I'd probably

5 quit by now, but I know that they need me. And if it's

6 not for our kids, it's for the next generation because

7 no family should ever be told their child has a

8 terminal illness and there's nothing you can do about

9 it. Thank you.

10 DR. WITTEN: Thank you very much. Our next

11 speaker is Alison Bateman-House. She's an Assistant

12 Professor in the Division of Medical Ethics at NYU

13 Grossman School of Medicine. She's co-chair of the

14 Working Group on Compassionate Use and Pre-Approval

15 Access, an academic group that studies ethical issues

16 concerning access to investigational medical products.

17 She has published and spoken extensively on how to best

18 handle requests for non-trial access to investigational

19 drugs and related ethical issues. And she's also

20 written and spoken frequently on the history and ethics

21 of using humans in research subjects and on clinical

www.transcriptionetc.com

308

1 trial accessibility. So welcome.

2

3 ETHICAL ISSUES IN PRODUCT DEVELOPMENT AND

4 SUSTAINABILITY FOR INDIVIDUALIZED THERAPIES-DR. ALISON

5 BATEMAN-HOUSE

6

7 DR. BATEMAN-HOUSE: Hi, everyone. I want to

8 thank the FDA for the invitation to speak today and I

9 want to thank Ms. Wood for that very illuminating

10 presentation. So there's no way I can follow that, so

11 I won't even try. The best I can probably do is

12 entertain y'all with my lack of ability to manipulate

13 mechanics. So we'll see how this works.

14 So we've heard numerous people say today, you

15 know, maybe the time has come that we need to come up

16 with a paradigm shift in this drug development

17 traditional model that we've heard of. You know, this

18 pre-clinical, Phase 1, Phase 2, Phase 3, it's too slow.

19 Maybe there's situations in which we need to modify it

20 somehow. And I think that's probably true, and we're

21 here to talk about that. But before we talk about

www.transcriptionetc.com

309

1 that, I want to just remind us how we got to where that

2 is. So let's see if I can move forward.

3 Okay. So in the 1940s, we basically said, you

4 know, let's do medicine scientifically, and we're going

5 to sort of disaggregate research and treatment. And

6 they may look very similar, the same products may be

7 involved, but they have different intentions. And as a

8 result -- sorry, I'm getting distracted by -- see, I

9 told you I'm bad at technology. I should just look up

10 here.

11 They have different intentions. So they may

12 involve, you know, similar procedures. They may

13 involve similar, you know, products, but the intention

14 is different. And here's the classic definition from

15 the 80s. Research talks about a class of activities

16 designed to develop or contribute to generalizable

17 knowledge. Whereas, practice is referring to a class

18 of activities designed solely to enhance the well-being

19 of an individual patient or client.

20 So when people go into research, we often hope

21 that there will be benefit to them, but it's not the

www.transcriptionetc.com

310

1 motivating factor behind research. And this has been

2 sort of a core disaggregation that happened, like I

3 said, in the '40s and has been systematized over time.

4 With this Phase 1, this Phase 2, Phase 3 paradigm that

5 we have now really came into place in the '60s, and it

6 said, research, it is a formal thing. It has a method,

7 and it has a procedure. And it's different from

8 treatment.

9 And as a result of that we sort of came up

10 with this traditional model of who is responsible for

11 what. Patients, your responsibility is to be treated,

12 be a good patient, do what your doctor tells you to do.

13 Doctors, your responsibility is to treat patients.

14 That is your patient in front of you. It is your job

15 to advocate for them and do whatever you can do to help

16 them. Researchers, your responsibility is to conduct

17 research and to get that generalizable data that will

18 move science and knowledge forward. Research subjects,

19 you are passive. Your role and responsibility is to be

20 researched upon. Funders, your role is to figure out

21 what research is promising, to vet it, to fund it. And

www.transcriptionetc.com

311

1 then, as we mentioned earlier, typically the

2 pharmaceutical companies are the ones who have come in

3 and said, you know, “We'll take it from here. We'll

4 take the most promising research and turn it into a

5 marketable medical product.”

6 Now, this started collapsing around the '80s.

7 We had activist patients, particularly in the context

8 of HIV/AIDS who said, “I don't want to be a passive

9 research subject. I want to have agency. This is not

10 working for me anymore.” And there are many, many

11 people today who say this is not working for me

12 anymore. And we've heard examples all throughout today

13 of patients or parents who have taken science into

14 their own hands and said, “In the effort to treat my

15 child, I need to get involved in the research world.”

16 So the traditional model is evolving. Just to

17 give some examples, you know, as we've heard today,

18 research may be intended primarily as therapy. If you

19 have a research endeavor that is anticipated to help

20 one patient, that's not research for the sake of

21 research. That's research as therapy.

www.transcriptionetc.com

312

1 As a result, your research subjects, they may

2 not me acting as passive research subjects. They're

3 saying “I'm a patient. I'm here to be cured or helped.

4 This is my therapeutic option.” Patients and

5 advocates, as we just heard a stunning demonstration

6 of, they may be the ones now who are picking what

7 science to fund, what science to push forward, what

8 science to really try to get out of the realm of theory

9 and actually into the lab.

10 This means that companies may be sidelined,

11 and not necessarily because they're being pushed out by

12 patients. I don't mean that. But as we mentioned

13 earlier, if there's no market incentive, companies are

14 basically leaving the space, and that's why these

15 parents or advocacy groups are coming in. And they

16 may not be sidelined, but they're playing a less

17 prominent role. And in some cases, they're almost even

18 like subcontractors.

19 And then there's the question that, you know,

20 we bring to the FDA of, well, what and how are we going

21 to approve something out of this? I mean, typically

www.transcriptionetc.com

313

1 speaking, if it's a N of 1, like really an N of 1

2 bespoke that no one other than person with the de novo

3 mutation is gonna use, you don't need anything

4 approved. But if there's something that we can

5 extrapolate form this like a platform technology, then

6 maybe there is something we can approve. What is it

7 and how are we gonna approve it? And will something be

8 brought to the market?

9 So these are all new questions that everyone

10 in this room is currently, you know, grappling with and

11 I just wanted to lay them out. And of course, anytime

12 you shift between paradigms, it's difficult and there

13 are complications. And so some of the questions that

14 arise as we're making this shift right now is I showed

15 you that traditional outlay of roles. And one of the

16 questions is who should be playing what roles?

17 So for example, if you have a laboratory

18 scientist, are those the people that we want making

19 treatment decisions for individual patients? Maybe

20 yes, maybe no, but it's not necessarily something that

21 they've done before or been trained for. And this is a

www.transcriptionetc.com

314

1 new reality that we need to be grappling with.

2 Funding. Funding has always impacted

3 decisions as to what's going to be developed, but is

4 funding now going to be impacting treatment decisions?

5 And I'll just give you an example from a patient

6 advocacy group that I work with. They funded the

7 clinical trial, and so the expectation was, you know,

8 well, we get to pick what patients go first. Who's the

9 first to get dosed? Who's the second to get dosed?

10 Who's the third to get dosed? And are we okay saying,

11 you know, this wouldn't be happening without you so

12 okay, you get to make that decision? Or is that sort

13 of a no go request?

14 So research and therapy traditionally have

15 been distinct, and there has always been rampant

16 confusion between where is this -- I think Dr. Witten

17 says, you know, sometimes there's a fuzzy line. And

18 there's a concept called therapeutic misconception.

19 Which, I used to work in cancer, and you saw

20 therapeutic misconception all the time with patients in

21 Phase 1 clinical trials. It used to be, before our

www.transcriptionetc.com

315

1 modern era of molecularly targeted cancer, Phase 1

2 clinical trials were not intended to be therapeutic.

3 They were intended to get dosage information and to

4 move the development of a molecule along. But the

5 patients enrolled in that Phase 1 trial were really not

6 anticipated to benefit.

7 Yet, we saw time and time again that if you

8 surveyed those patients and ask why they were

9 participating in the clinical trial they would say,

10 “You know, I'm hoping to get something out of it.” So

11 there's been this ongoing confusion about, am I

12 participating in research? And if so is that therapy?

13 Is it not therapy? And it's been even more complicated

14 when the same health care provider has been the

15 investigator and the clinician. And the patient or

16 parent is like, “Well, am I being recommended to go

17 into this trial because they think it'll help me or

18 because they're the PI of this trial and they need

19 people?”

20 So this has been an ongoing issue for decades.

21 And of course, now we're getting to the situation where

www.transcriptionetc.com

316

1 research and treatment are getting even closer together

2 and, in some cases, becoming completely inseparable.

3 And that's what we're looking at in some cases with

4 some of the individualized therapeutics.

5 Although I was struck today as we heard

6 different stories from cancer vaccines to gene therapy

7 to gene editing, that there are distinctions between

8 them. You know, you can't make a one size fits all

9 statement here. But regardless, in general, we're

10 seeing the situation in these individualized

11 therapeutics where there is this sort of merger of, you

12 know, from the expectation of the participant, is this

13 research or is this treatment?

14 And that has implications for those of us who

15 are in the field. Those of -- implications for, you

16 know, the companies, the clinicians, the researchers,

17 the academic medical centers. So if something is

18 experimental but it's intended as treatment, how do we

19 handle that? Because traditionally we've had a model

20 where those have been disaggregated. And it has legal

21 implications. It has regulatory implications. It has

www.transcriptionetc.com

317

1 ethical implications.

2 And just to single out one, IRBs. So if

3 you’re doing research, you have to go through an IRB.

4 You have to get ethical review of your research.

5 That's very different from what happens in treatment.

6 In treatment, there's still the idea of you

7 need to have informed consent, but you don't go through

8 an IRB. Your paperwork that you fill out to enroll in

9 a clinical trial is very different, normally speaking,

10 from the paperwork you fill out to have, like, your

11 gall bladder removed. So you -- we have these concepts

12 that, yes, there must be informed consent. But how we

13 actually formalize them, change has been different

14 depending on which one of these realms you're in. And

15 so we need to figure out really where we are.

16 So a number of people have pointed out today

17 that some of these individualized therapies have been

18 given under the rubric of expanded access, also known

19 as compassionate use, in some cases called the eIND.

20 And I spent a lot of time in this field, so I just

21 wanted to make some comments about it. So we mentioned

www.transcriptionetc.com

318

1 IRBs a second ago. IRBs are involved in both research

2 and EA.

3 But the level of oversight is very different.

4 So if you have an IRB involved in our research, there's

5 gonna be multiple rounds of review probably, looking at

6 every line of the protocol and trying to decide how is

7 this gonna be advertised, and who's gonna be recruited,

8 and how do we make sure they understand what they're

9 getting into, and what sort of data are we gonna return

10 to them? The IRBs on expanded access basically say,

11 does this seem reasonable and is there some piece of

12 paper that we can hand somebody, either the patient or

13 the family member, to have them sign off that they

14 understand that this is experimental?

15 The levels of oversight are completely

16 different. So there are just practical ramifications

17 to trying to figure out -- we need to figure out which

18 one of these paradigms we're working in. And, just as

19 I'm saying right now, what does it matter?

20 So you know, pick a paradigm and just move

21 forward. Say it's research, say it's expanded access,

www.transcriptionetc.com

319

1 say it's treatment, say whatever, it doesn't matter.

2 It does matter because we need clarity about the

3 procedures. We need clarity both within a particular

4 institution or within a particular multi-center

5 initiative or what have you. We need to make sure that

6 patients understand what's being proposed, why and what

7 the possible risk and benefits are.

8 And of course, the way we currently do that is

9 through an informed consent discussion that is

10 memorialized with informed consent form. But we need

11 to know what that form looks like, and we need to try

12 to choose which one of these to use to do that

13 properly. And we need to make sure that the

14 stakeholders involved understand their

15 responsibilities.

16 So I just wanted to go briefly over this. And

17 expanded access when you're talking about, you know,

18 sort of, like, your classic idea of an unapproved drug,

19 the request to do, you know, use this unapproved drug

20 outside of a clinical trial has to be initiated by the

21 physician. Of course, the patient may be the one who

www.transcriptionetc.com

320

1 initiates this conversation with the physician, but you

2 have to have the physician on board. And the physician

3 is the one who is in charge of reaching out to the

4 company who is developing this new product and saying,

5 “May we please use this outside of the clinical trial.

6 Here is the patient, here's why I want to use it, and

7 here's why I can't use it in a trial.”

8 And if the company says no, that's basically

9 the end of the story. This is where we see social media

10 campaigns and whatnot trying to make companies change

11 their minds. But generally speaking, if a company says

12 no, that's the end of the story.

13 If a company says yes, this is where the FDA

14 gets involved to look over the proposal, make sure that

15 there's no obvious safety concerns, see if there are

16 any amendments that they think need to be added to the

17 proposal. And of course, this is where the IRB gets

18 involved. So this is your sort of ladder that you have

19 to go through to do expanded access for a single

20 patient.

21 And I just want to note that really the

www.transcriptionetc.com

321

1 gatekeeper in this situation is the company. Again, if

2 the company says no, that's basically the end of the

3 story. The FDA cannot say to the company, “You must do

4 this.” The IRB cannot say to the company, “You must do

5 this.” It's the company that's the gatekeeper. And I

6 want to point this out because, in the situations that

7 we've been hearing today, the roles of companies have

8 been changed, if not minimized or completely removed.

9 And we're really talking about things that are

10 happening in the academic center.

11 So is the role of gatekeeper now being taken

12 over by this investigator, and, if so, does that

13 investigator know that they are now the gatekeeper? Do

14 they want to be the gatekeeper? Are they comfortable

15 being the gatekeeper? You could say there is no

16 gatekeeper; everyone gets what they want. But I don't

17 think that's a sustainable model, and we probably

18 shouldn't advocate it.

19 But if the investigator doesn't want to be the

20 gatekeep do we say, “Okay, FDA, now you're the

21 gatekeeper? Every time someone wants to use one of

www.transcriptionetc.com

322

1 these individualized products outside of a clinical

2 trial, you really need to do the due diligence and

3 decide yes or no.” And it's up to the FDA to decide if

4 they're comfortable with that role or not, but that's

5 an additional burden over what they've currently been

6 asked to assume.

7 Or we could say to the IRB, “Hey, before

8 you've kind of done a rubber stamp. You know you've

9 looked at this and said, ‘Is it reasonable?’ and okay

10 make sure that there's a piece of paper for someone to

11 sign. But now we're in a new paradigm, and you really

12 need to be involved.” I don't know. Any of these are

13 possible, but there's a question. And we need to

14 figure out what to do.

15 I just want to point out that I do have a

16 concern about the idea of this investigator being the

17 gatekeeper, although it might seem like the obvious

18 choice, simply in that I'm concerned that having

19 proximity to a patient might make that investigator

20 make decisions that are a little bit too close for

21 comfort. And the sort of paradigmatic example I'm

www.transcriptionetc.com

323

1 thinking of is if you have a patient who is

2 deteriorating in front of you. Is there some point

3 where the investigator would say, “Gosh, in an ideal

4 world we would, you know, do some more work on this?

5 I'm not 100 percent comfortable, but we can't wait

6 anymore.” And maybe that's okay if it really is an N

7 of 1 and this is the only patient that it's gonna

8 impact.

9 But I don't think that's okay if there are

10 other patients out there that we're gonna be trying

11 this intervention on, and hopefully we'll be collecting

12 data from this first experiment. And to the idea that

13 someone might be -- jump the gun a little bit, that

14 makes me concerned. And of course, there's also the

15 point where if that patient or that patient's family or

16 that patient's community is the one funding the

17 investigator, is there even more of this potential

18 conflict of interest?

19 And, you know, if you can think of these

20 problems happening theoretically then they're probably

21 gonna happen in real life. And so is there something

www.transcriptionetc.com

324

1 we can do to prevent foreseeable issues? Say, we're

2 fine with the investigator being the one making these

3 decisions, but let's come up with some rules of

4 engagement.

5 So just to repeat, we need to decide is this

6 research? And if it is research, we need to modify the

7 way that we think about these. And we need to train

8 IRBs so they understand what they're looking at. And

9 we need to come up with some way of deciding what

10 experimental procedures we really are happy with and

11 whatnot. Or if we say this is clinical care, fine.

12 That's fine. I'm okay with that, but we need to,

13 again, come up with rules of engagement.

14 And one of the questions I have is, you know,

15 do we say any licensed M.D. in the country should be

16 able to do this or only certain M.D.s, only in certain

17 settings, only with certain oversight? And if we are

18 gonna say there are some sort of rules of engagement,

19 who's gonna develop them and who's gonna enforce them?

20 All right. So then to get into the bread and butter of

21 ethics concerns I always have to talk about justice.

www.transcriptionetc.com

325

1 So one of the things that I'm concerned about

2 here -- and this is not to say we should not do this.

3 I absolutely think we should do this, but these are

4 questions we need to ask. How do we justify extensive

5 use of resources to benefit only one person or maybe

6 very few people?

7 And I think everyone in the room today has

8 been hitting upon the same theme of, you know, we

9 collect data from those N of 1s, and we find some way

10 to have that data push us forward in ways that will

11 help more patients. So I think that's great that we're

12 all on the idea of leveraging findings to help wider

13 numbers of patients. But we need to figure out how to

14 do that. We must plan for it, and it must be something

15 that we say is non-negotiable, not something that, you

16 know, after we do some cases, we'll figure out how to

17 go back and do a reanalysis of the data. I don't think

18 that's acceptable. I think we need to say right from

19 the start, as we're building our plans, here's how

20 we're gonna do this.

21 Enhancing access, decreasing obstacles, and

www.transcriptionetc.com

326

1 costs. I think just last week there was a meeting,

2 maybe here at FDA -- I don't know -- for Rare Disease

3 Week. And there was a panel on individualized

4 therapeutics, and there was a patient who had received

5 phage therapy. And she talked very movingly about how

6 she was receiving care in Richmond, Virginia. And she

7 had a very bad intractable infection and came up with

8 the idea of phage therapy.

9 And her physician either was unable or

10 unwilling to do it, and so she had to go to Yale to get

11 access and how she literally thought she might die on

12 the train because it was just too much to ask for her

13 to do it. And she did it. And it worked, and that's a

14 success story. But it's also a cautionary tale because

15 I'm deeply concerned about the idea that we are, you

16 know, not thinking about access from the get-go and

17 trying to figure out how to minimize those barriers as

18 much as possible.

19 We live in a country that has unjust access to

20 health care, and so I understand when I'm saying we

21 need to enshrine justice as a core principle in access

www.transcriptionetc.com

327

1 to research that seems a little bit odd because we

2 don't have it in access to just normal clinical care.

3 But I'm aspirational like that. What can I say?

4 You know, so as we're building a phage bank or

5 as we're developing consortium, I just am encouraging

6 people to invest the time, the money, and the planning

7 up front to try to figure out how to minimize barriers

8 to patients both in real time and downstream. And

9 also, I have an international point here -- I'm not

10 sure we've talked internationally yet today -- about is

11 it possible to harmonize regulations now from the get-

12 go so that once something is successful in the United

13 States we can very easily translate it to, say, Canada

14 or other countries where people will be wanting to try

15 these?

16 More justice concerns. We've talked about

17 this numerous times today in terms of how do we decide

18 how much information we need to acquire before we use a

19 product on a patient. And, you know, I don't have a

20 magic bullet answer for this. This is an ongoing

21 question anytime you have a novel intervention. We're

www.transcriptionetc.com

328

1 trying to balance earlier access and the possible

2 benefit it offers with the risk that comes with getting

3 something, you know, earlier before all the testing has

4 been done.

5 So my only recommendation here is that we need

6 to embed frequent evaluations of safety. It can't be

7 something where we do, you know, we're gonna do 20

8 people, and then we're gonna do a, you know, a post hoc

9 analysis. I think we should be looking at this in real

10 time as we go along and manage risk cautiously.

11 And then, last but not least, I just wanted to

12 say, the thing that really gives me heartburn about all

13 of this is that, as we are doing this novel, like,

14 amazing science for certain patients, there are other

15 patients who are being told, “I'm sorry you can't get

16 access because it hasn't gone through a mouse model

17 yet, or it hasn't gone through a primate model yet.” Or

18 “the Phase 1 was only enrolling 20 patients, and

19 they've already enrolled those 20 patients. so you're

20 gonna have to wait until a Phase 2 trial opens.” It is

21 hard for me to understand how we are gonna justify to

www.transcriptionetc.com

329

1 other patients who are not in this, like, exclusive,

2 individualized therapy category about why they still

3 need to observe a status quo and rules that are, you

4 know, being turned and modified in this particular

5 context.

6 So that's the thing that keeps me up at night.

7 And in the meantime, the thing that makes me really

8 happy is the fact that we're having, like, amazing

9 science that's gonna help patients. So I want the

10 amazing science to help patients to go forward, but I

11 want us to be aware of all the challenges that it

12 presents for us and to be proactive about addressing

13 them. Thank you.

14 DR. WITTEN: I'd like to next introduce Dr.

15 P.J. Brooks. He's a program director at the NCATS

16 Office of Rare Diseases and Research. He received his

17 Ph.D. in neurobiology from the University of North

18 Carolina at Chapel Hill and completed a postdoctoral

19 fellowship at the Rockefeller University. Since

20 joining NCATS and the Office of Rare Diseases, Dr.

21 Brooks has been working on accelerating clinical trials

www.transcriptionetc.com

330

1 in rare diseases by moving beyond one disease at a time

2 approaches. Examples include the development of

3 therapeutics that target shared molecular mechanisms,

4 underlying multiple rare diseases, platform

5 technologies for delivery of nucleic acid therapeutics,

6 and the implementation and recommendations from the

7 NCATS Cures Acceleration Network regarding the

8 acceleration of gene therapy clinical trials. Thank

9 you.

10 BEYOND ‘ONE DISEASE AT A TIME:’ ACCELERATING CLINICAL

11 TRIALS OF GENETIC THERAPIES BY GROUPING RARE DISEASE

12 PATIENTS ACCORDING TO UNDERLYING DISEASE MECHANISM -

13 DR. PHILIP J. BROOKS

14

15 DR. BROOKS: Thank you Celia and thank you to

16 my FDA colleagues for the invitation to participate in

17 this meeting. It's really exciting and it's something

18 that we think about a lot. And I'm very happy to

19 participate. And I'll be focusing on, more generally,

20 the idea of trying to go beyond one disease at a time

21 and how we can accelerate clinical trials of genetic

www.transcriptionetc.com

331

1 therapies by grouping rare disease patients according

2 to underlying mechanism.

3 This is my standard federal government

4 disclosure slide. And here you see the basic problem

5 that I think we're all trying to address is the rapidly

6 increasing number of disorders with a known molecular

7 basis, due in large part to DNA sequencing. And this

8 is likely to continue. And the big problem is that, at

9 the present time, we've only about 600, 500 or 600 or

10 so with therapy. And at the rate we're going, as my

11 director, Chris Austin, said, it's gonna take about

12 2,000 years to get treatments for every one of these

13 diseases, and that's just too darn long. And if we're

14 gonna do something about this we don't need sort of

15 minor tweaks to the process. We need some pretty

16 fundamental changes in the way we think about these

17 diseases and the way we design clinical trials.

18 And there's another aspect to this as well

19 that sometimes doesn't get appreciated, but I think

20 Jill Wood kind of hit on it. And that has to do with

21 this issue of the different types of rare diseases. So

www.transcriptionetc.com

332

1 this is a slide taken from a recent publication by

2 Orphanet, and we're looking at, within rare disease,

3 there's different prevalence, you know, the highest and

4 the lowest. And if you're thinking about the

5 percentage of all rare disease patients, the majority

6 of them are these high prevalence diseases, right?

7 That makes sense, about 70 percent of the patients.

8 But if you think about the number of diseases, it's a

9 very small fraction. Way on the other side, the low

10 prevalence diseases, the Sanfilippo, you know, III C,

11 D, et cetera, you've got a very small number of

12 patients but about 3,000 or so diseases.

13 And then, if we're gonna develop these

14 treatments according to a standard model, the diseases

15 of commercial interest are these ones for obvious

16 reasons. But who's ever gonna do anything about these

17 diseases here, these 3,000 diseases? If we're gonna do

18 this one at a time I'm not sure we're gonna get to any

19 of them. So we have to really reevaluate how we

20 approach this problem, particularly because, as I'll

21 get to later, when we're talking about some of these

www.transcriptionetc.com

333

1 diseases and monogenic diseases, we really do have

2 therapies and treatments that have a pretty high prior

3 probability of success. Oops. Wrong way. Okay.

4 So I think it comes down to the old lumpers

5 and splitter distinction that we're all familiar with

6 from different facets of life. This was taken from a

7 paper written by Victor McKusick, the famous

8 geneticist, many years ago. And he was talking about

9 limpers and -- lumpers and splitters in the context of

10 nosology. As you can tell by the way he drew these

11 different pictures, he was a big fan of the splitters

12 and not so much the lumpers. And maybe that made sense

13 for the point he was trying to make; I honestly don't

14 know. But with all due respect, I think in this day

15 and age we've got to be lumpers wherever we can. And

16 we've really got to focus on the commonalities across

17 diseases rather than what makes them different.

18 And the really, I think, wonderful opportunity

19 here is in the area of monogenic diseases. And you

20 could say this is perhaps the biggest lump of all, at

21 least within personalized or individual therapies.

www.transcriptionetc.com

334

1 Because, when you talk about monogenic diseases, these

2 are diseases that relate from mutations in a single

3 gene. And that means we know what the problem is, and

4 we know at least what some solutions are, which is

5 quite different than many other diseases.

6 So we have gene therapy which could deliver a

7 normal version of that mutant gene into the relevant

8 cell types of the patient and then, more increasingly

9 now, genome editing where you can -- need to deliver

10 genome editors or enzymes into these cell populations

11 to correct that disease-causing mutation in the

12 patient's cells. Particularly here we're talking about

13 somatic cells. That's what we focus on in the United

14 States. We're not -- we don't do germline editing in

15 the United States.

16 And so the idea then -- so the real challenge

17 for both of these is to deliver these treatments to

18 enough target cells at the right time in development of

19 disease progression to potentially treat, cure, or even

20 prevent some of these diseases. And this is really

21 fundamentally true for all monogenic diseases. And the

www.transcriptionetc.com

335

1 good news here is that we've actually made some

2 progress in being able to do this. As Guangping Gao

3 talked about adeno-associated virus or AAV, these

4 really are effective vectors to deliver genes into

5 cells.

6 They have an excellent safety record in humans

7 to date, clinical success stories to approved products,

8 and we see a lot of pre-clinical success stories. If

9 you go to the American Society for Gene and Cell

10 Therapy meetings, there's a lot of people curing a lot

11 of mice of a lot of genetic diseases. It works quite

12 well. But when you go to develop these into the

13 clinic, we run into this one disease at a time

14 approach.

15 And quite often you see the focus on the more

16 common rare diseases. And this is slow, inefficient

17 and results in duplications of efforts across different

18 programs. It costs animals, time, and money and

19 particularly the time and money in some cases of the

20 parents who are trying to develop these therapies, of

21 which there isn't very much. And there's this obvious

www.transcriptionetc.com

336

1 bias towards the more common rare diseases. So what

2 makes sense would be to do -- to start these clinical

3 trials for multiple diseases at a time using the same

4 platform vector and that should increase the efficiency

5 and reduce the time of clinical trial start up, would

6 make sense.

7 So we're gonna try to test that specifically

8 in a program we call the Platform Vector Gene Therapy

9 or PaVe-GT project. And this is a collaborative effort

10 between our office, Office of Rare Diseases Research at

11 NCATS, and other collaborators at NCATS, in particular

12 the Therapeutics Development Branch led by Don Lo and

13 strategic alliances led by Lili Portilla, as well as

14 colleagues from NHGRI and NINDS who will be working

15 with us on the clinical trial. Do you have some water?

16 So to be clear, this is a pilot project where

17 we're gonna be doing essentially a public platform

18 vector gene therapy trial at the NIH Clinical Center

19 involving all investigators from NIH. And the idea is

20 to move forward with -- ultimately, towards clinical

21 trials for gene therapy for four rare genetic diseases

www.transcriptionetc.com

337

1 together, each of which are of no commercial interest.

2 So these are -- these -- the very far end of that graph

3 I showed before. We use the same AAV vector, the same

4 route of administration, the same serotype, use the

5 same production purification methods because we've

6 heard from our FDA colleagues many, many times that the

7 process is the product. And the only thing that will

8 be different are the therapeutic gene constructs for

9 the different diseases. And the question is to what

10 extent can we increase the speed and efficacy of

11 clinical trials startup by really trying to maximize

12 this explicit platform vector-based approach.

13 But the other thing I think that's gonna be

14 different about this is that we intend to do this

15 publicly and make all of the data, including the

16 biodistribution data, the toxicology data, all of our

17 communications with the FDA up to and including the IND

18 submissions that will hopefully get approved -- we

19 intend to make that public and publicly available so

20 that all these documents and things can be used by

21 others perhaps even in a cut and paste manner. And in

www.transcriptionetc.com

338

1 doing this and thinking about this approach, honestly,

2 we had in mind people like Jill and all the parents

3 that we meet who are trying to figure out how to do

4 this by themselves. You know, it's got to be hard

5 enough having a child with a rare genetic disease.

6 But then to expect them to become

7 entrepreneurs and drug developers, and we think how can

8 we provide help for some of these individuals? And I

9 wish there was more we can do, but this is one approach

10 that we think has potential benefit. And essentially,

11 the idea here is that if we're going to do these one

12 disease at a time for each vector we would make, for

13 each gene, for each disease, we go through each of

14 these steps in parallel, one right after the other.

15 And that takes time and money, and time is an issue.

16 But the question is, if you group them all together,

17 can we utilize the fact that we're doing everything in

18 the same pathway to avoid having to do, perhaps the

19 biodistribution studies or some of the other steps, and

20 reduce the amount of time to clinical trial startup?

21 And so here's sort of where we are on this.

www.transcriptionetc.com

339

1 We've got the collaborating investigator Carsten

2 Bonnemann from NINDS, and Chuck Venditti from NHGRI.

3 And we'll be working with two neuromuscular diseases

4 and two rare organic acidemias using AAV9 for all four.

5 And we're undergoing -- proof of concept studies and

6 mouse models in human cells.

7 And I say we're gonna let you know about our

8 communication with the FDA, and I can tell you that we

9 had a communication with the FDA a few months ago and

10 talked to them about this. And it went quite well,

11 better than I kind of anticipated. They were quite

12 good about the idea and I think were all supportive

13 about being as transparent as possible. And we’ll be

14 anticipating our initial INTERACT meeting with the FDA

15 later this year. And the key challenge we're facing,

16 which I guess is one that everybody is facing and

17 probably led to the meeting we had a few weeks ago, is

18 how do we get AAV vector made for the clinical trials?

19 Because we run into the same problem that everybody

20 else does. So we are working on that as well.

21 And then earlier, Peter Marks, mentioned the

www.transcriptionetc.com

340

1 effort that we've been involved with working with them,

2 as well as the FNIH, to develop a more broad public-

3 private partnership for some of these individualized

4 therapies and specifically focusing on AAV gene

5 therapy. And it's a pleasure to be working with them

6 and look forward to continue doing so. And I just

7 wanted to point out that there are some actual

8 parallels between our PaVe-GT effort and this other

9 public-private partnership that is in progress that we

10 hope can ultimately be leveraged.

11 In both cases, there has to be some decision

12 about the serotype that's going to be used for the

13 different diseases. We're choosing a single one.

14 Perhaps there'll be multiples here. I put question

15 marks on all these because there's still some questions

16 about how our -- this public-private partnership is

17 gonna work, but I think there's some clearly -- clear

18 issues that'll need to be addressed. We're doing all

19 of our work at the NIH Clinical Center.

20 I might point out, in part because we want to

21 utilize that resource, but also because we're so

www.transcriptionetc.com

341

1 committed to making all of the data publicly available

2 that we felt that the most efficient way to do it is by

3 having everyone be a government employee. If we had a

4 commercial company involved or even academic medical

5 centers, we might have to deal with some of the

6 intellectual property issues. So having it all done

7 within the NIH kind of avoids that problem.

8 In -- oops. But ultimately in this effort we

9 might be involving many clinical sites. As I said,

10 we're gonna make all of our communications public. And

11 what we had kind of hoped, actually, is that some of

12 these documents and things might ultimately spill over

13 and benefit this potential public-private partnership.

14 The single manufacturer is, of course, key.

15 But one might consider a consortium here, and,

16 you know, we chose four rare diseases for specific

17 reasons and largely due to the availability of diseases

18 and the investigators at the NIH. But here in this,

19 whatever, effort, I think a big question is gonna be

20 how do we choose and determine what diseases that would

21 be under consideration? But I think both of these

www.transcriptionetc.com

342

1 things are kind of moving forward towards a day in

2 which we can really make access to these treatments a

3 lot more available for a lot -- much larger numbers of

4 patients and families and hopefully take the burden off

5 of people like Jill and other parents.

6 So then also the next phase, if you will, of

7 the way to treat genetic diseases is genome editing.

8 And I just want to briefly touch upon a program that

9 we're involved within at NIH. This is a program funded

10 by the NIH Common Fund, part of the Office of the

11 Director. And it's on somatic cell genome editing.

12 And it's coordinated by NCATS, Chris Austin, myself,

13 and in association with many other program directors

14 across the NIH.

15 And the goals of this program are to lower the

16 barriers for new genome editing therapies by testing

17 genome editing reagents and delivery systems and better

18 animal models. These are not specific disease models

19 but rather animal models created to allow us to detect

20 genome editing in different cell types to maximize a

21 broad utility, a big focus on testing unintended

www.transcriptionetc.com

343

1 biological effects. And I should say, unintended

2 biological effects specifically in human cells and

3 human cell systems for the reasons that Peter

4 mentioned, that the human genome is special. We also

5 have some interest in monitoring these cells in vivo.

6 We have some -- the biggest focus of the

7 program is finding ways to deliver genome editors to

8 different cell types. There's also a small part on

9 increasing the genome editing repertoire, sending

10 genome editing enzymes, and of course a coordinating

11 center. And to give you a sense of the breadth of the

12 program, this is the number of awards. The total

13 budget of the program is around $180 million. And you

14 can see that by far the majority of the awards --

15 almost half of them are focused on the delivery systems

16 because that, as we see, is the biggest challenge.

17 There are some cells and tissues we can deliver to

18 pretty well but many that we can't at all, and that's

19 really what the focus is on.

20 And if you want to learn more about it, here's

21 the website. And the way we see this focused --

www.transcriptionetc.com

344

1 getting into the IND enabling process is kind of

2 illustrated here, that this program will not

3 specifically be funding any clinical studies, but

4 rather what we think about is filling gaps. And there

5 might -- one gap might be for a specific disease a need

6 to be able to deliver genome editors to a particular

7 cell type. But another gap actually that we focus on,

8 and would really like to have some impact on, is the

9 gaps in the regulatory process. That would allow our

10 FDA colleagues to be able to regulate these products

11 more effectively. And indeed, when we were developing

12 this program and as we are going through it, we have

13 close communication with people in FDA CBER to try to

14 maximize that potential.

15 So finally, let me turn to addressing the

16 question that FDA asked us to address which is what

17 kind of opportunities and possibilities might work in

18 the future to make some of these approaches scalable

19 and sustainable. And I have some thoughts about that.

20 So one would be to adapt the approach they use now in

21 the development of vaccines, when one is producing a

www.transcriptionetc.com

345

1 new vaccine using a new strain. I should say that this

2 is not an idea I came up by myself. This was

3 originally brought to me by one of my colleagues, Mike

4 Cirillo at NCATS, and I've heard Peter talk about it

5 also.

6 But the basic idea is that, when someone's

7 first gonna set up a vaccine production facility, the

8 FDA would review -- would have to consider all of the

9 aspects of setting up the process: the manufacturing

10 facility, CMC potency assays, all kinds of things in

11 addition to specific strains. But once a system is

12 ongoing and producing, if from one year to the next

13 you're just simply switching strains, then the FDA

14 review can just focus on what's different, what's new,

15 which in this case would be the new strain. And I'm a

16 little imprecise about this because I've never

17 regulated a vaccine, but I think you kind of get the

18 idea.

19 And so we could take the same basic principle

20 and apply it to gene therapy. When you're originally

21 developing a gene therapy product, the FDA's gonna have

www.transcriptionetc.com

346

1 to consider all these things, the manufacturing and the

2 whole, sort of, regulatory process. But the idea and

3 the hope is that once we're making the same vector, and

4 in the same manufacturing facility and holding all

5 these things the same and simply changing the

6 therapeutic transgene, the FDA review can focus on

7 this. And that could streamline the process. And not

8 surprisingly, that's basically the idea we're trying to

9 test in PaVe-GT.

10 But I think the real exciting option has to do

11 with genome editing. And late last year this

12 publication came out from David Liu, who is funded by

13 our consortium in part, a new genome editor that can

14 carry out editing without creating double strand

15 breaks. And the notable thing about this prime editing

16 effort -- this prime editing enzyme is that this single

17 enzyme could, in principle, correct almost 90 percent

18 of known genetic disease-causing mutations. And that

19 really seems like a potentially exciting platform.

20 And so if you imagine -- and again, obviously,

21 I'm thinking about the future here. These are perhaps

www.transcriptionetc.com

347

1 forward-thinking statements. But if you think about

2 the production utilization of such a biologic, you can

3 end up with a single biologic that would be of

4 potential therapeutic relevance to almost 90 percent of

5 genetic diseases.

6 And once that would be approved, then, if you

7 just want to add additional diseases to it, the only

8 things that would be different are these guide RNAs

9 that would direct the location of the editor within the

10 genome. Guide RNAs of course are oligonucleotides, and

11 oligonucleotides are regulated by the Center for Drugs.

12 But the good news is, of course, a lot of excitement

13 going on in this area with the oligonucleotide

14 therapies for the rare genetic diseases.

15 And I'm sure you've all heard about the work

16 by Tim Yu on the development of Milasen, and obviously

17 the FDA, CBER, and CEDR are able to communicate on

18 this. So it doesn't seem like an insurmountable

19 problem. And so if you get back to this sort of

20 optimized situation in the future, we’d be looking at

21 perhaps a single biologic. And then the guide RNAs

www.transcriptionetc.com

348

1 would be assessed under really a streamlined process

2 that takes into account the platform capacity of

3 oligonucleotides to be able to see them as a class of

4 molecules that would optimize the toxicology assessment

5 of those as well. And, you know, thinking about the

6 future of treating monogenic disease, this seems to me

7 to be at least an aspirational idea of where we might

8 want to go.

9 And so I think I'll just kind of stop there

10 and summarize that, for monogenic diseases, gene

11 therapy and gene editing have clear and obvious

12 therapeutic potential for many monogenetic diseases.

13 This one disease at a time approach that we're doing

14 now is not going to address these low prevalence

15 diseases of no commercial interest, despite the fact

16 that the biological rationale of treating those

17 diseases is just as good as the common diseases. And

18 that does not seem acceptable. That's a major reason

19 why we need to do something different. And so we need

20 radically different types of clinical trials and

21 regulatory platforms to bring gene therapy and gene

www.transcriptionetc.com

349

1 editing therapies to all the patients who might benefit

2 from them.

3 Oh, and just one last thing. I think we've

4 been always talking about individual therapies, and I

5 understand why we're saying that. But I was telling

6 you at the beginning, there's lumpers and there's

7 splitters. And when you talk about individual diseases

8 you're focused on splitting.

9 And I think -- I happened to come across this

10 from a publication in Stat News. And I like the idea

11 of industrializing personalization because I think

12 that's sort of what we're talking about, how to take

13 making individualized therapies into some sort of an

14 industrial process. So I guess I'll leave you with

15 that as well as our NCATS contact information. Thank

16 you very much.

17 PANEL DISCUSSION WITH Q&A

18

19 DR. WITTEN: Okay. Thank you. I'm gonna ask

20 the speakers and the -- to take their seats at the

21 panel. And also, I'm going to ask Dr. Chip Schooley

www.transcriptionetc.com

350

1 and Dr. Julienne Vaillancourt. Dr. Schooley was

2 introduced in the last session, but I just want to

3 introduce Captain Julie Vaillancourt, whom many of you

4 know. She's an officer in the U.S. Public Health

5 Service, and she's the Rare Disease Liaison for the

6 Center for Biologics Evaluation and Research and

7 coordinates our rare disease program.

8 Thank you for joining. We'll take questions

9 from the audience, but, in the meantime, I have a few

10 questions to start off the discussion. I’d like to ask

11 the two panel members who just joined us first. So

12 I'll start with Julie. This session has been about

13 collaborations, ethics, and stakeholder roles. And I

14 wonder if you can comment on collaborations at FDA,

15 CBER, and the rare disease area that supports

16 product development?

17 CAPT. VAILLANCOURT: Absolutely. Actually, we

18 have a really rich -- oh, thank you. At CBER, we have

19 a very rich collaborative environment when it comes to

20 our focus on advancing development of biological

21 products for rare diseases. We have collaboration

www.transcriptionetc.com

351

1 internally in CBER. We actually, as part of our rare

2 disease program, have a Rare Disease Coordinating

3 Committee that meets on a monthly basis. It's

4 comprised of representatives from each of the offices

5 in CBER: our product offices, our Office of

6 Epidemiology and Biostatistics, our -- and others.

7 And also, we collaborate extensively across

8 the agency with the Office of Orphan Products

9 Development, OOPD, and with CDER's rare diseases

10 program. And I also want to say a newer entity in the

11 last two or more years is the Patient Affairs staff in

12 the Office of the Commissioner. And they are very

13 instrumental in helping to facilitate making sure that

14 the patient voice is heard and that there are ways and

15 many mechanisms to engage patients with each of the

16 centers. And we continue to work with these different

17 groups across the agency, and we're developing new

18 collaborations every day.

19 Since I've been the rare disease liaison in

20 September of 2015, I've been participating in a Rare

21 Disease Council that is headed by the Office of Orphan

www.transcriptionetc.com

352

1 Products. And again, it's an effort that brings

2 representatives that work in -- on rare disease focused

3 development from across the agency. We meet

4 periodically. We share information, best practices.

5 Sometimes we bring in outside speakers. There's also a

6 new Rare Disease Round Table that was started by

7 Theresa Mullen in the Office of Drugs that is engaging

8 some outside stakeholders as well.

9 We have -- I'm going on and on, but you can

10 see it's very rich. We also coordinate and have a rare

11 disease cluster with our European colleagues. It's a

12 rare disease cluster that is headed by CDER's Rare

13 Diseases Program, but a number of us from CBER

14 participate. And sometimes we are asked to have

15 discussions with CBER regulated products and issues

16 with our EMA colleagues. And Health Canada has more

17 recently joined in those monthly discussions.

18 And we also coordinate with our outside

19 stakeholders, our external stakeholders, such as the

20 National Organization for Rare Disorders, NORD. We

21 have been part of their planning committee for a number

www.transcriptionetc.com

353

1 of years. And we also have some newer cooperative

2 agreements with them like our -- someone from our

3 Office of Biostatistics and Epidemiology is working on

4 a collaborative project with NORD and with a patient

5 advocacy group on a natural history study and the use

6 of a mobile app.

7 So there's lot of exciting work going on. And

8 I'll have to say we're making room for the whole topic

9 of today's meeting of individualized therapeutics. And

10 it's, for example, we've mentioned -- a few people have

11 mentioned today about the rare disease meeting that

12 took place last Monday or -- yeah, it was last Monday.

13 It's gone by so fast.

14 Anyway, that was headed by Dr. Janet Maynard

15 from the Office or Orphan Products. However, it was a

16 collaborative team effort, and there was representation

17 from CBER and from CDER. And the afternoon was all

18 focused on individualized therapeutics. And, you know,

19 there are other examples, so I hope I've given you a

20 sense of the breadth and depth of collaboration that

21 CBER is involved in, as we really work toward the

www.transcriptionetc.com

354

1 development of biologics for patients with rare

2 diseases.

3 DR. WITTEN: Okay. Thank you. I'm gonna take

4 a question from the audience.

5 MS. HESTERLEE: Hi, so it's Sharon Hesterlee

6 from the Muscular Dystrophy Association. So I'm very

7 interested in this problem of the ultra-rare diseases.

8 At MDA, you know, we hear a lot about Duchenne and SMA

9 and ALS, but the majority of the over 45 diseases we

10 cover are ultra-rare. So this is a problem that I

11 think about a lot and that keeps me up at night. And

12 I've also spent the last three-and-a-half years working

13 in industry in gene therapy and heading gene therapy

14 projects, in charge of budgets for gene therapy

15 projects. So this is kind of comments for P.J.

16 I'm really interested in this effort to create

17 a platform approach. But the caution I would give you

18 is that this idea of using the same serotype, or the

19 same capsid and vector, the same route of

20 administration, you know, same manufacturing

21 techniques, these are things that companies are already

www.transcriptionetc.com

355

1 doing. I mean, they're already doing that to try to

2 get those economies of scale. And I'm gonna tell you,

3 your savings in money are very, very small. Savings in

4 time, also pretty small. So I don't know that this

5 platform approach and this idea of this platform

6 approach at the pre-clinical, early clinical phase is

7 really gonna be that big of a time or cost savings.

8 I do think where you could see more savings

9 are doing things like doing a platform trial approach.

10 The problem is these transgenes matter, so there's only

11 so much you can do to sort of combine your efforts at

12 the pre-clinical stage. You can have tox related to

13 different transgenes and expression of transgenes. I

14 just wanted to make that point that I think companies

15 are already doing this.

16 They're already standardizing those things.

17 They're already standardizing their manufacturing

18 assays. They're doing all of those things. It's not

19 enough. Your budgets are still $20, $30 million, even

20 for an ultra-rare disease. So I think it's just

21 something to consider that that may not be as tractable

www.transcriptionetc.com

356

1 a target in trying to reduce costs.

2 DR. WITTEN: Thank you. Are there comments?

3 Do you have comments on this?

4 DR. BROOKS: Yeah. I think -- I'm sure there

5 are companies doing that, but I think that information

6 isn't made available. So we would want to make it

7 available to the whole community so it's not kept

8 within a company. And I think that certainly there

9 are many cost drivers in this, and we don't expect that

10 this effort by itself will, you know, reduce that.

11 I think our focus of the AAV manufacturing

12 meeting that we had was to try to find ways to reduce

13 the cost. But improving not just even the pre-clinical

14 stuff, but even as we think about the clinical trial

15 design within PaVe-GT, I do think we'll also be looking

16 for ways to increase the efficiency of the process.

17 But to try to learn what we can do and also to make it

18 public so everybody can benefit from it is a major

19 aspect of this goal. But certainly there's -- there

20 are -- there's more to do than that. I would agree.

21 DR. WITTEN: Thank you. I'd like to address a

www.transcriptionetc.com

357

1 question to Dr. Schooley. So Dr. Schooley, this is

2 about developing products. So you described in your

3 talk the collaboration of iPATH and of the phage

4 referral network so that, if a patient came in need of

5 a treatment, the group would collectively search their

6 inventory to see if there was phage available that

7 would benefit them. But that that only -- that was not

8 true for the -- I think you said it was the minority of

9 the patients who came that you were able to find

10 something. So I'm wondering what type of collaboration

11 or what type of effort do you think would be needed and

12 by whom in order to be able to develop phages for -- so

13 that no infectious disease is left behind, so to speak?

14 DR. SCHOOLEY: Over the short term, the

15 problem is the biology. You actually physically have

16 to have the organism in -- the bacterium in the same --

17 on the same plate or in the same liquid medium as the

18 phage candidate you want to use, which requires you to

19 have -- to disseminate that organism to whatever labs

20 or groups have libraries of phages that target that

21 organism. So right now, the limitation is how many

www.transcriptionetc.com

358

1 groups have large enough libraries that you can

2 practically get a given patient's organism out in time

3 to be able to help the patient.

4 Having more comprehensive libraries that could

5 be screened more easily would be act- -- and methods by

6 which you could screen them quickly would be great.

7 Down the longer term, if you had -- if we could by AI

8 learn to predict from AI what bacteria could be

9 attacked by which phages, you could actually do it with

10 whole genome sequencing. We're a long way from that

11 because there are more variables than equations these

12 days. But that would be the -- down the road, I think,

13 a very important approach.

14 One could also envision situations in which

15 phages were engineered to have a broader host range, in

16 essence become more like antibiotics and less like

17 phages. What you would be giving up there is the

18 specificity of phages from the standpoint of the

19 microbiome and the other advantages of the laser like

20 approach. And you would also begin to see, if you had

21 widespread use of phages with engineered phage with

www.transcriptionetc.com

359

1 broad host range, they would behave like antibiotics do

2 in the hospital. You would begin to have phage

3 resistant organisms that would then behave the same way

4 antibiotic resistant organisms do.

5 So I think, over the short term, larger phage

6 banks that could be searched more easily and production

7 facilities that were able to take that burden off the

8 hands of academic laboratories and produce phages in a

9 more standardized way would increase the throughput.

10 The sources of funding for those really haven't yet

11 been identified.

12 DR. WITTEN: Thank you. Question from the

13 audience?

14 MR. THAKUR: Yes, I'm Neil Thakur from the ALS

15 Association. So I had a question for P.J. about the

16 model that you're talking about and the ultimate vision

17 of success. And so I think what you were saying is the

18 idea is that you would get a manufacturing process

19 approved by the FDA. And when it comes time to bring

20 on a new disease or a new application of the

21 technology, the FDA review would be expedited. And so

www.transcriptionetc.com

360

1 what I'm trying to understand then is does that mean

2 that the clinical center or the facility that's doing

3 all this manufacturing would then become the hub for

4 AAV9 for these ultra-rare applications? Or would

5 somehow -- could this model be expanded or exported to

6 other facilities as well? So what's your -- what's the

7 step after this project, in other words?

8 DR. BROOKS: So I guess I'm trying to be clear

9 because I'm talking about two different things. The

10 PaVe-GT is one and then the individualized therapies

11 that we're working with FNIH and FDA CBER -- is that

12 what you're referring to?

13 MR. THAKUR: No. You had a slide where you

14 talked about on one side you had here's what the FDA is

15 gonna review in great detail, and then ultimately, when

16 you bring on a new thing, it'll happen faster. And I'm

17 -- I think that was the PaVe-GT ultimate thinking. But

18 I'm not clear on what the final status that you're

19 trying to drive to, how you see the manufacturing, and

20 the FDA, and the NIH all working together.

21 DR. BROOKS: Yeah. I think there's different

www.transcriptionetc.com

361

1 levels and different projects. But I think the idea in

2 part was to make the regulatory process easier for the

3 FDA. When we're adding on -- if they want to add on --

4 we want to add on a new disease, if the vector

5 manufacturing is one they've seen before and the

6 biodistribution has been seen before that we wouldn't

7 have to repeat that. And that could increase our

8 clinical trial startup and make the regulatory path

9 easier.

10 I don't think I would imply that we're gonna

11 get FDA to approve a manufacturing process. I don't

12 see -- it's not obvious to me how that would work, but

13 I think it would be something like having a -- you

14 know, using the same process over and over again and

15 having the focus just be on what's different.

16 MR. THAKUR: And that'll be the clinical

17 center doing that going forward in long term?

18 DR. BROOKS: No. I don't think in long term.

19 I think optimally in long term we'd want to expand

20 this. And I think expanding beyond the PaVe-GT, our

21 pilot project, would ultimately hope- -- potentially be

www.transcriptionetc.com

362

1 the FNIH public-private partnership, and that would not

2 be limited to the NIH Clinical Center.

3 MR. THAKUR: Thank you.

4 DR. WITTEN: Before we take the next question,

5 I just want to clarify. So I -- what P.J. said is

6 correct. I mean, that is a shared, you know, his

7 description of it. We don't license processes. But if

8 we learn from the process or what happens and we learn

9 from our review and we learn from the science, that'll

10 facilitate continuing development, which is I think

11 what the goal of the program is. So that's -- I think

12 we're in agreement about what we think will happen, the

13 benefit could be. Yes?

14 MR. HORGAN: Rich Horgan from Cure Rare

15 Disease. One of the things tying together the ethical

16 and the stakeholder issues I think we may have been

17 overlooking a bit is the role of the payer in this. So

18 in the last two months we've had conversations with

19 chief medical officers of two of the biggest payers or

20 insurance companies in the United States. They are

21 aware of the development of customized therapeutics.

www.transcriptionetc.com

363

1 And the current mechanism for reimbursement is one that

2 is not at all conducive to reimbursing customized

3 therapies. More of a comment than a question, but an

4 urge to consider the payer perspective as you're

5 designing these, whether it's a platform trial or other

6 thinking both at the NIH as well as the FDA, because I

7 think, at the end of the day, if we can prove that we

8 have efficacious and safe custom drugs for one or two

9 patients, that's certainly great. But it's not

10 sustainable if we don't have payers on our side and

11 supportive of this approach.

12 So sort of urge thinking and more thought

13 surrounding that area because these certainly aren't

14 cheap, especially when we get to larger volume AAV

15 deliveries like with a Duchenne or another

16 neuromuscular disease. It's not an eye, and it's not

17 as privileged as the CNS in being compartmentalized to

18 some degree, but, you know, would urge some thinking

19 around that area.

20 DR. WITTEN: Thank you. Are there comments

21 from the panel on this topic?

www.transcriptionetc.com

364

1 DR. BATEMAN-HOUSE: I want to thank you for

2 bringing that up. I've been thinking a lot about

3 payers today. And I don't remember who said it, so I'm

4 not calling out names. But someone today said

5 something about, you know, well, if we had an add on

6 therapy that was safe, and even it was only a little

7 effective, why wouldn't we do it? And I immediately

8 thought because payers won't pay for it. So I think

9 you can't lose sight of that, especially when you're

10 thinking about access downstream.

11 So it's one thing to say in this interim

12 period, we don't need to worry about payers. But

13 whenever I talk with a company, and in this case

14 whenever I talk with an academic center or anybody

15 doing novel development, I would say think downstream.

16 Who you plan on using this product, and what evidence

17 do you need to get? At what level of certainty do you

18 need to convince payers to make that actually happen?

19 Because it's one thing to get FDA approval; it's

20 another thing to go through that other set of

21 gatekeepers which are payers.

www.transcriptionetc.com

365

1 DR. WITTEN: Thank you. I think --

2 DR. BROOKS: Oh --

3 DR. WITTEN: Sorry.

4 DR. BROOKS: I just -- I'll just make one

5 point. I think, I mean, I certainly agree about the

6 payer point. I think one of the other efforts that we

7 have in the Office of Rare Diseases Research is to try

8 to understand the cost of all these rare diseases on

9 our current health care system. Because when the

10 payers are going to be thinking about this, it's the

11 cost of paying for the therapy compared to the cost of

12 not having the therapy. And understanding the current

13 costs of all these rare diseases on our medical system

14 is -- we don't really have good data on that for a

15 variety of reasons, in part because of the difficulty

16 of the lack of ICD codes for some of these diseases.

17 So we have quite an effort going on at NCATS to come up

18 with a good estimate of what we call the cost of rare

19 because that will help the payer consideration.

20 DR. WITTEN: Do we have a question from the

21 online viewers?

www.transcriptionetc.com

366

1 THE OPERATOR: Yes, we do. The topic is

2 ethics.

3 DR. WITTEN: It's hard to hear you.

4 THE OPERATOR: The topic is ethics. And the

5 question is, in what I'm hearing, clinical trials are

6 very much being spoken of as treatment. How much

7 concern is there about research subjects or patients

8 clearly understanding and giving consent to early

9 trials that have not yet established safety?

10 DR. BATEMAN-HOUSE: So I think that was the

11 point that I was trying to make is that, traditionally,

12 we have said that there is research that should not be

13 thought of as therapy and that there is a high bar that

14 is expected to be cleared in terms of the informed

15 consent process that is asked of when a patient goes to

16 participate versus we had a much lower informed consent

17 process to participate in a therapeutic endeavor. And

18 the example I gave was surgery. So you know, you sign

19 a one page very small consent form to have your gall

20 bladder removed versus a very complex consent form to

21 participate in a clinical trial.

www.transcriptionetc.com

367

1 That's the model that we all grew up with.

2 That's the model that has always been said is a best

3 practice. But the question is are we at a point where

4 that model need transforming? And, in some cases, it

5 may be that it does need transforming.

6 If a truly bespoke therapy is being done on

7 one person even though it is experimental, it's not

8 necessarily research anymore. So we need to come up

9 with some understanding of how to navigate that divide.

10 And then the other point that I had hoped to make is

11 that that doesn't mean that that distinction is gonna

12 collapse across the board. So there is still going to

13 be areas where there is a divide between research and

14 therapy, and we still need to make sure that patients

15 in those contexts understand that going into this

16 clinical trial is not necessarily a therapeutic

17 endeavor.

18 And so there's a possibility of having mixed

19 messages about, you know, yay, all research is

20 treatment when that's really not true. And how do you

21 make those clarifications clear to potential research

www.transcriptionetc.com

368

1 subjects? I'm sorry. I'm looking at you because

2 you're the one that asked the question. I should be

3 looking -- I don't know who I'm supposed to be looking

4 at. The camera. Hello, camera.

5 So I just -- I'm very concerned in terms of

6 understanding, transparency, and expectations that we

7 need to be very clear about what is status quo, what is

8 different, and, if there are differences, why there are

9 differences and, if people are being held to the status

10 quo per se, why that is as opposed to it just being,

11 like, we like this disease and we don't like this

12 disease, or this disease has more engaged patients

13 versus this disease doesn't? I think we need to be

14 more clear as to why we're acting in certain ways in

15 certain paradigms.

16 DR. WITTEN: Thank you. I'm going to take the

17 two questions from the people who are already lined up

18 and then take the chair's prerogative to ask the final

19 question before closing the session.

20 MS. NOSRATIEH: Thank you. This is Anita

21 Nosratieh from FasterCures. This is a question going

www.transcriptionetc.com

369

1 back to the payer considerations and thinking about

2 this truly end-to-end access. Do you think, P.J., it

3 would be possible to incorporate CMS into the pilot

4 that you guys are spearheading between NIH, FDA? Just

5 seems like a natural, kind of, inclusion.

6 DR. BROOKS: I’m looking over at Peter. Okay.

7 Yeah. So Peter's going to address that later. Thank

8 you.

9 MS. BLACK: Hello. Lauren Black with Charles

10 River Laboratories and ex-CBER. I'm interested to see

11 the analogies between the current personalized medicine

12 and where we stand today in terms of monogenic

13 diseases. Within the context of monogenic diseases, I

14 think it's more like surgery, as Alison pointed out,

15 where the patient comes in. You can do an analysis

16 that's equivalent to saying, okay, the artery is

17 bleeding. We know what's wrong with the patient.

18 We know that they need a specific enzyme to be

19 replaced, or they need a certain gene replaced or

20 knocked out. We know exactly what's wrong with those

21 patients. So for that subset, this is very surgical.

www.transcriptionetc.com

370

1 It seems as if the payer would take a more surg- --

2 investigational surgery type approach to saying okay,

3 we're gonna replace that gene. We can measure that.

4 We can say if the initial drugs are working and have a

5 pharmacodynamic response.

6 And then say that yes, there's a blurred line

7 between treatment and research, but we can see that the

8 gene that was missing is now producing that protein.

9 And we can detect that protein in the blood and the

10 CSF. That seems to be a lot more clear than trying to

11 treat a disease that we didn't understand the cause or

12 had multiplicities of causes.

13 Here we have a much more specific thing that

14 we're looking to accomplish, so why shouldn't the lines

15 be blurred? Because as soon as we can detect the

16 replaced protein in the person, we know that we're much

17 closer to actually remediating their condition. So I

18 think this is actually a place where we can make a sea

19 change because we can see what's wrong, and we can see

20 how to fix it.

21 DR. WITTEN: Well, I think it's -- we're all

www.transcriptionetc.com

371

1 optimistic that this approach will work. So I think

2 we'll have to see what happens with this.

3 DR. BATEMAN-HOUSE: Can I say something about

4 it?

5 DR. WITTEN: Oh, sure.

6 DR. BATEMAN-HOUSE: So I'm not sure there was

7 a question there, and, if there was, I didn't get it.

8 So sorry if I don't answer correctly. But I just

9 wanted to say -- one thing that I just want to make

10 sure is clear, when I am saying that the intention may

11 be therapeutic but something being done is still

12 experimental and hence we need to figure out how to

13 deal with the informed consent and other problems of

14 that nature, is I want to share a conversation I had

15 with Dr. Timothy Yu who has been mentioned several

16 times.

17 So he had a terminally ill child who had a de

18 novo mutation of Batten's disease and was able to say,

19 I think I can come up with a customized therapy that

20 will help this child based on a platform from Spinraza

21 that I can make some alterations and use it to

www.transcriptionetc.com

372

1 potentially help this child. The thing that he was

2 concerned about he told me going into this was no one

3 had done this before, and there was a possibility that

4 by infusing this experimental product into this child,

5 who was blind, was not really able to communicate, and

6 was obviously headed towards an early demise, he could,

7 yes, actually, intervene in that trajectory, but he

8 could have other potentially unintended side effects.

9 And the one that he was concerned about was

10 awakening or reinvigorating some part of the brain that

11 would allow her to experience pain and thought I really

12 don’t want to give this intervention to a child that

13 may, yes, prolong her life but may also make her

14 current state of being worse. So I think even though

15 there's a very sort of cut and paste mentality, like,

16 of course this is gonna work -- it's very logical and

17 it could work -- it's still experimental. We don't

18 know what's gonna happen.

19 And even though we could say it's more of an

20 experimental therapy than research because the intent

21 is to help this person, you can't lose fact of the --

www.transcriptionetc.com

373

1 sight of the fact that it is experimental. And you

2 have to have that understanding going into it and make

3 sure that there is the informed consent and make sure

4 that everybody understands, you now, we don't know

5 what's gonna happen here. Maybe after a couple

6 iterations in a couple different settings, we'll have a

7 better basis for being able to make predictions but not

8 at first.

9 DR. WITTEN: Thank you. So my last question --

10 and I'm gonna ask everyone on the panel. And I'll

11 start with Jill, if that's okay -- is -- of course you

12 don't know the question, so I guess it's maybe not fair

13 to ask if it's okay. Is, if there's one thing -- so we

14 obviously at CBER are looking at these questions very

15 carefully as to what we can do to facilitate the

16 process or what we need to look at, what our next steps

17 should be to try to benefit patients and benefit

18 product development.

19 And so I just would like to know from each of

20 you, if you have any thoughts, if you do have any

21 thoughts on what would success look like for us just

www.transcriptionetc.com

374

1 for the next year? I don't mean 10 years, you know, 20

2 years success, but just, if we could accomplish

3 something in the next year, what would it look like?

4 If you have a comment on that.

5 MS. WOOD: Well, I think what P.J. is

6 suggesting is profound, and it's absolutely amazing.

7 And if you could pull it off, you'd be pulling off a

8 decade of success right there. Something very easy I

9 would say, a success is to identify those ultra-rare

10 diseases and uber rare diseases that really need --

11 have a need for mouse models and registries and natural

12 history studies and help facilitate those --

13 development of those things by either working with

14 their patient organizations or trying to figure out an

15 in-house way of doing that for all.

16 DR. WITTEN: Thank you. Next. Alison.

17 DR. BATEMAN-HOUSE: I think the fairest answer

18 to say is I don't know.

19 DR. WITTEN: Okay.

20 DR. BATEMAN-HOUSE: But I guess I would just

21 say I really think it's important to be transparent in

www.transcriptionetc.com

375

1 these ongoing conversations and also, in terms of -- to

2 the extent that CBER is starting to lean certain ways,

3 to divulge that as soon as possible because it sounds

4 like there's a lot of people waiting for some sort of -

5 - certainly as to whether they're on the right path or

6 not. And the sooner that they can feel some sense of

7 assurance that, you know, maybe we don't have a final

8 guidance yet, but we see that there's a wind blowing

9 this way, that would be helpful.

10 DR. WITTEN: Thank you.

11 DR. BROOKS: So I think one of the most

12 exciting things that I see is the effort that involved

13 with Peter and FNIH to develop this public-private

14 partnership and really test a very different way to do

15 gene therapy clinical trials for bespoke therapies. I

16 don't know that we can pull it off and get it started

17 within a year. But I think if we can do that, to have

18 the FDA leadership involved in an effort like this

19 seems like a very different approach for the FDA

20 leadership. And I think it's really wonderful and

21 exciting for all of us who are participating in it and

www.transcriptionetc.com

376

1 I think, you know, for the whole community. So…

2 DR. WITTEN: Thank you.

3 DR. SCHOOLEY: You know, we're working with

4 often rare diseases. And we should try to learn how to

5 generalize our knowledge base, so we don't have to

6 discover the same thing over and over again in each

7 specific clinical indication, so learning how to

8 generalize with the skepticism you need about over

9 generalization, at the same time -- and to focus our

10 resources on moving the field forward rather than just

11 repetitively doing the same thing over and over again,

12 just say this is the way we develop this therapeutic.

13 In other words, connecting the dots in a more -- among

14 these efforts in a more cross-fertilizing way will help

15 us move forward. And I think that, at the end of the

16 day, if we can do that, we'll help in both these kinds

17 of diseases and how we approach other diseases as well.

18 The innovation that we've seen today in these

19 approaches, I think, is really breathtaking, and that

20 innovation needs to be balanced with the care that we

21 approach human engagement in research.

www.transcriptionetc.com

377

1 But also, we need to make sure that what we

2 learn on platform that has a lot of similarities to

3 others is shared so we didn't have to discover the same

4 thing over and over again. And parallel efforts, that

5 holds everybody back because of proprietary needs that

6 put these people at risk over and over again who

7 studied the same thing and slows the field. So

8 generalization early in the process helps everyone,

9 particularly our patients, so early sharing of

10 approaches, platforms, techniques. We're all here for

11 the same thing, and there's plenty of room for

12 innovation in any given field is what I would argue.

13 DR. WITTEN: Thank you. And as usual FDA gets

14 the last word, so…

15 CAPT. VAILLANCOURT: Thank you. Well, I think

16 today is an extremely important day. We're starting a

17 public dialogue about this critically important area

18 and in a way of -- to go forward without having all the

19 burden on patients and parents, such as Jill and others

20 out there. And I think it's so important that we keep

21 the dialogue going, that we don't lose momentum.

www.transcriptionetc.com

378

1 And I'd like to agree with what P.J. said,

2 and, being from FDA, I think this announcement of this

3 public-private partnership is very, very exciting. So

4 be great if we could convene again and see some -- hear

5 about the status of what's happening with that public-

6 private partnership, also to get an update on how the

7 NIH program is going. I mean, these are all really

8 exciting initiatives -- but also to hear more from our

9 stakeholders.

10 We're just so thrilled that everybody is here.

11 For those of you in the room and everybody on the

12 phone, the whole intention was to get the stakeholders

13 together today and to hear from everyone. So again,

14 keep the dialogue going, keep the momentum going.

15 DR. WITTEN: Thank you. I'd like to thank the

16 panel and the speakers. And next I'm going to turn it

17 over to Dr. Peter Marks, our Center Director for -- to

18 wrap it up.

19

20 WRAP UP AND CLOSING REMARKS: DR. PETER MARKS

21

www.transcriptionetc.com

379

1 DR. MARKS: So thank you everyone in the room

2 and online who had stuck with us for the full day. I

3 think rather than summarizing each of the sessions, I

4 think what I just want to say is, I think what really

5 came through pretty clearly is that I think we all see

6 the compelling need to make headway here in these

7 individualized or bespoke therapies. And I think we --

8 in each of the sessions we have these building blocks

9 that we can build upon, whether it's on the

10 manufacturing, the non-clinical aspects, the clinical

11 aspects, or patient access to these things.

12 Just to back up to try to address some of this

13 because the issue of how do you pay for these things

14 has come up. So I think there are lots of different

15 ways to think about this. But the way I would think

16 about this for at least for the reason why we're very

17 interested in a public-private partnership is I think

18 we're in a time of transition here. We're in a time of

19 transition where -- there was a model several years ago

20 where every rare disease would be commercially viable

21 somehow because you could charge enough for the gene

www.transcriptionetc.com

380

1 therapy.

2 I think it's -- I'm oversimplifying it, but I

3 think what we've realized is -- and what companies have

4 realized is that commercial viability lies beyond many

5 of the diseases that we're talking about today, which

6 means we have to find some other way to fill in that

7 gap. I will tell you an opinion. This is not the

8 opinion of the United States Government Health, and

9 Human Services or the Food and Drug Administration of

10 the United States of America.

11 This is my own opinion that, 10 to 15 years

12 from now, this issue will be fixed because there will

13 be commercial viability for very rare bespoke therapies

14 inasmuch as I think much of this will be essentially

15 dealt with by having machines that can -- this will be

16 a device issue. Many of these gene therapies will be

17 made potentially on non-viral platforms or by

18 mechanisms that don't require the kind of setup that

19 they currently do. And we will have had a lot more

20 experience about what you can leverage.

21 But for this interval of the next 5 to 10

www.transcriptionetc.com

381

1 years, I think we have to find a way to get these

2 therapies to individuals in need. I don't know that

3 we're gonna find a way to get payers to pay for them,

4 at least in the short term. But what I do think is

5 there's a lot of good will going between companies,

6 non-profit organizations, and, for that matter,

7 government wanting to collaborate with them to find

8 ways to try to make these therapies available to those

9 in need.

10 There's gonna be a lot of ethical issues, a

11 lot of prioritization issues that'll have to be worked

12 through in this. But I think that's the goal of these

13 public-private partnerships is to try to find a way

14 forward. And ultimately, the reason why this is so

15 important, at least to me, is that this is a case

16 where, if we can get it right for these very small

17 numbers of patients, ultimately the entire field of

18 gene therapy is bound to benefit. So it's one of these

19 things that start small and local and then go more

20 globally.

21 And just so that I just mention that we didn't

www.transcriptionetc.com

382

1 concentrate here today on the discussion of the more

2 global issues for gene therapy. But part of the reason

3 for getting it right here is that, if we can take care

4 of these products on a small scale here, hopefully, we

5 can have a global framework so that patients around the

6 globe will benefit from their development. It really

7 would be a shame if we spent the time developing these

8 here, and then they're not accessible -- you know, it

9 would be really sad for a Sanfilippo type C or a type

10 III C or D patient here not to get something and not to

11 have a patient in Asia or in Africa benefit from that

12 same advance that we've made.

13 So ultimately, I think we're gonna go back

14 from this. We will do what FDA likes to do is we will

15 cogitate for a while. I think we do understand that

16 ultimately the way we try to put forth our thinking is

17 in guidance. Hopefully that will be forthcoming at

18 some point in the future and in the not too distant

19 future.

20 And we'll also continue to work with our

21 partners at FNIH and NCATS to try to move forward this

www.transcriptionetc.com

383

1 public-private partnership. Is success assured? No,

2 it's not. But I think it's certainly worth a try

3 because there's a lot of good will there. There are a

4 lot of patients in need of these therapies, and I do

5 think we have to try to do something differently that

6 will try to get us there.

7 We have actually -- just to answer another

8 question, we have actually spent some time with

9 business folks and with companies talking about

10 economies of scale. And there probably are some

11 economies of scale to be had here in part by using

12 excess capacity, in part by reusing certain aspects of

13 files and et cetera. It's not gonna ever be cheap, but

14 we do think this is something that could hopefully lead

15 us to be able to more efficiently get there for

16 patients.

17 So with that I just want to close by saying I

18 really want to thank all of the speakers today,

19 particularly Ms. Wood who I really think really shared

20 a very compelling picture of what it's like to deal

21 with this type of situation from a variety of aspects.

www.transcriptionetc.com

384

1 And to all of you for -- thank you for coming today and

2 for really caring about this issue. And I'd also echo

3 something that Julie Vaillancourt said which is that we

4 look forward to continuing the dialogue with everyone.

5 And with that I have two last things to do.

6 One of them is to once again to thank Leslie Haynes and

7 Gopa Raychaudhuri for really planning an incredibly

8 excellent workshop. So let's give them a round of

9 applause. And then I told Gopa I'd give her the last

10 word. So here she goes. You're good. Okay. With

11 that, thank you very much. Okay. Thanks again,

12 everyone.

13 [MEETING ADJOURNED]

www.transcriptionetc.com